1
|
Peregud DI, Shirobokova NI, Kvichansky AA, Stepanichev MY, Gulyaeva NV. Purmorphamine Alters Anxiety-Like Behavior and Expression of Hedgehog Cascade Components in Rat Brain after Alcohol Withdrawal. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1938-1949. [PMID: 39647823 DOI: 10.1134/s0006297924110087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/13/2024] [Accepted: 08/06/2024] [Indexed: 12/10/2024]
Abstract
Disturbances in the Hedgehog (Hh) signaling play an important role in dysmorphogenesis of bone tissue and central nervous system during prenatal alcohol exposure, which underlies development of fetal alcohol syndrome. The involvement of Hh proteins in the mechanisms of alcohol intake in adults remains obscure. We investigated the role of the Hh cascade in voluntary ethanol drinking and development of anxiety-like behavior (ALB) during early abstinence and assessed changes in the expression of Hh pathway components in different brain regions of male Wistar rats in a model of voluntary alcohol drinking using the intermittent access to 20% ethanol in a two-bottle choice procedure. Purmorphamine (Hh cascade activator and Smoothened receptor agonist) was administered intraperitoneally at a dose of 5 mg/kg body weight prior to 16-20 sessions of alcohol access. Purmorphamine had no effect on the ethanol preference; however, rats exposed to ethanol and receiving purmorphamine demonstrated changes in the ALB during the early abstinence period. Alcohol drinking affected the content of the Sonic hedgehog (Shh) and Patched mRNAs only in the amygdala. In rats exposed to ethanol and receiving purmorphamine, the level of Shh mRNA in the amygdala correlated negatively with the time spent in the open arms of the elevated plus maze. Therefore, we demonstrated for the first time that alterations in the Hh cascade induced by administration of purmorphamine did not affect alcohol preference in voluntary alcohol drinking. It was suggested that Hh cascade is involved in the development of anxiety after alcohol withdrawal through specific changes in the Hh cascade components in the amygdala.
Collapse
Affiliation(s)
- Danil I Peregud
- Serbsky National Medical Research Center for Psychiatry and Drug Addiction, Ministry of Health of the Russian Federation, Moscow, 119034, Russia.
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Nataliya I Shirobokova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Aleksei A Kvichansky
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Mikhail Yu Stepanichev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
- Research and Clinical Center for Neuropsychiatry, Moscow City Health Department, Moscow, 115419, Russia
| |
Collapse
|
2
|
Dylag KA, Wieczorek-Stawinska W, Burkot K, Drzewiecki L, Przybyszewska K, Tokarz A, Dumnicka P. Exploring Nutritional Status and Metabolic Imbalances in Children with FASD: A Cross-Sectional Study. Nutrients 2024; 16:3401. [PMID: 39408368 PMCID: PMC11478469 DOI: 10.3390/nu16193401] [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: 09/14/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Malnutrition is a significant concern in paediatric populations, particularly among children with neurodevelopmental disorders such as foetal alcohol spectrum disorder (FASD). This study aimed to examine macronutrient and micronutrient imbalances and assess the nutritional status of a group of patients with FASD. METHODS This study involved an analysis of the serum levels of key nutrients in a group of children diagnosed with FASD. Macronutrients and micronutrients were measured to identify any imbalances, including vitamin D, B12, E, A, albumin, and serum protein, among others. RESULTS The study found a high prevalence of vitamin D deficiency among the patients. Additionally, elevated serum concentrations of micronutrients such as vitamin B12, E, and A were observed in 8%, 7%, and 19% of patients, respectively. Macronutrient imbalances were noted, including high levels of albumin and serum protein, indicating a possible metabolic disturbance. Unexpectedly, high rates of hypercholesterolemia were observed, raising concerns about an increased risk of metabolic syndrome in this population. CONCLUSIONS These findings suggest that the principal issue among patients with FASD is an altered metabolism rather than nutritional deficiencies. Potential causes of these abnormalities could include oxidative stress and changes in body composition. The results underline the need for further research to better understand the unique nutritional challenges in children with FASD and to guide the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Katarzyna Anna Dylag
- Department of Pathophysiology, Jagiellonian University Medical College, 31-121 Krakow, Poland
- St. Louis Children Hospital, 31-503 Krakow, Poland (A.T.)
| | | | | | | | | | | | - Paulina Dumnicka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Poland
| |
Collapse
|
3
|
Zeng X, Cai Y, Wu M, Chen H, Sun M, Yang H. An overview of current advances in perinatal alcohol exposure and pathogenesis of fetal alcohol spectrum disorders. J Neurodev Disord 2024; 16:20. [PMID: 38643092 PMCID: PMC11031898 DOI: 10.1186/s11689-024-09537-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
The adverse use of alcohol is a serious global public health problem. Maternal alcohol consumption during pregnancy usually causes prenatal alcohol exposure (PAE) in the developing fetus, leading to a spectrum of disorders known as fetal alcohol spectrum disorders (FASD) and even fetal alcohol syndrome (FAS) throughout the lifelong sufferers. The prevalence of FASD is approximately 7.7 per 1,000 worldwide, and is even higher in developed regions. Generally, Ethanol in alcoholic beverages can impair embryonic neurological development through multiple pathways leading to FASD. Among them, the leading mechanism of FASDs is attributed to ethanol-induced neuroinflammatory damage to the central nervous system (CNS). Although the underlying molecular mechanisms remain unclear, the remaining multiple pathological mechanisms is likely due to the neurotoxic damage of ethanol and the resultant neuronal loss. Regardless of the molecular pathway, the ultimate outcome of the developing CNS exposed to ethanol is almost always the destruction and apoptosis of neurons, which leads to the reduction of neurons and further the development of FASD. In this review, we systematically summarize the current research progress on the pathogenesis of FASD, which hopefully provides new insights into differential early diagnosis, treatment and prevention for patents with FASD.
Collapse
Affiliation(s)
- Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
4
|
González-Flores D, Márquez A, Casimiro I. Oxidative Effects in Early Stages of Embryo Development Due to Alcohol Consumption. Int J Mol Sci 2024; 25:4100. [PMID: 38612908 PMCID: PMC11012856 DOI: 10.3390/ijms25074100] [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: 03/03/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Alcohol, a widely consumed drug, exerts significant toxic effects on the human organism. This review focuses on its impact during fetal development, when it leads to a spectrum of disorders collectively termed Fetal Alcohol Spectrum Disorders (FASD). Children afflicted by FASD exhibit distinct clinical manifestations, including facial dysmorphism, delayed growth, and neurological and behavioral disorders. These behavioral issues encompass diminished intellectual capacity, memory impairment, and heightened impulsiveness. While the precise mechanisms underlying alcohol-induced fetal damage remain incompletely understood, research indicates a pivotal role for reactive oxygen species (ROS) that are released during alcohol metabolism, inciting inflammation at the cerebral level. Ethanol metabolism amplifies the generation of oxidant molecules, inducing through alterations in enzymatic and non-enzymatic systems responsible for cellular homeostasis. Alcohol consumption disrupts endogenous enzyme activity and fosters lipid peroxidation in consumers, potentially affecting the developing fetus. Addressing this concern, administration of metformin during the prenatal period, corresponding to the third trimester of human pregnancy, emerges as a potential therapeutic intervention for mitigating FASD. This proposed approach holds promise for ameliorating the adverse effects of alcohol exposure on fetal development and warrants further investigation.
Collapse
Affiliation(s)
- David González-Flores
- Department of Anatomy, Cell Biology and Zoology, Faculty of Medicine and Health Sciences, University of Extremadura, 06006 Badajoz, Spain
| | - Antonia Márquez
- Department of Anatomy, Cell Biology and Zoology, Faculty of Medicine and Health Sciences, University of Extremadura, 06006 Badajoz, Spain
| | - Ilda Casimiro
- Department of Anatomy, Cell Biology and Zoology, Faculty of Sciences, University of Extremadura, 06006 Badajoz, Spain;
| |
Collapse
|
5
|
He C, Guo Z, Zhang H, Yang G, Gao J, Mo Z. Identification and validation of methylation-CpG prognostic signature for prognosis of hepatocellular carcinoma. Aging (Albany NY) 2024; 16:1733-1749. [PMID: 38244582 PMCID: PMC10866447 DOI: 10.18632/aging.205454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024]
Abstract
Epigenetic biomarkers help predict the prognosis of cancer patients and evaluating the clinical outcome of immunization therapy. In this study, we present a personalized gene methylation-CpG signature to enhance the accuracy of survival prediction for individuals with hepatocellular carcinoma (HCC). Utilizing RNA sequencing and methylation datasets from GEO as well as TCGA, we conducted single sample GSEA (ssGSEA), WGCNA, as well as Cox regression. Through these analyses, we identified 175 oxidative stress and immune-related genes along with 4 CpG loci that are associated with the prognosis of HCC. Subsequently, we constructed a prognostic signature for HCC utilizing these 4 CpG sites, referred to as the HCC Prognostic Signature of Methylation-CpG sites (HPSM). Further investigation revealed an enrichment of immune-related signal pathways in the HPSM-low group, which demonstrated a positive correlation with better survival among HCC patients. Moreover, the methylation of the CpG sites in HPSM was found to be closely linked to drug sensitivity. In vitro experiments tentatively confirmed that promoter methylation regulated the expression of BMPER, one of the CpG sites within HPSM. The expression of BMPER was significantly correlated with cell death in the oxidative stress pathway, and overexpression of BMPER effectively inhibited HCC cell proliferation. Consequently, our findings suggest that HPSM is an independent predictive factor and holds promise for accurately predicting the prognosis of HCC patients.
Collapse
Affiliation(s)
- Chunmei He
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
- Chandi Precision Medical Technology, Foshan 528000, Guangdong, China
| | - Zehao Guo
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
- Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, China
| | - Hao Zhang
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
- Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, China
| | - Ganqing Yang
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jintao Gao
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
- Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, China
| | - Zhijing Mo
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, China
- Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, China
| |
Collapse
|
6
|
Goncalves-Garcia M, Hamilton DA. Unraveling the complex relationship between prenatal alcohol exposure, hippocampal LTP, and learning and memory. Front Mol Neurosci 2024; 16:1326089. [PMID: 38283699 PMCID: PMC10811250 DOI: 10.3389/fnmol.2023.1326089] [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: 10/22/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Prenatal alcohol exposure (PAE) has been extensively studied for its profound impact on neurodevelopment, synaptic plasticity, and cognitive outcomes. While PAE, particularly at moderate levels, has long-lasting cognitive implications for the exposed individuals, there remains a substantial gap in our understanding of the precise mechanisms underlying these deficits. This review provides a framework for comprehending the neurobiological basis of learning and memory processes that are negatively impacted by PAE. Sex differences, diverse PAE protocols, and the timing of exposure are explored as potential variables influencing the diverse outcomes of PAE on long-term potentiation (LTP). Additionally, potential interventions, both pharmacological and non-pharmacological, are reviewed, offering promising avenues for mitigating the detrimental effects of PAE on cognitive processes. While significant progress has been made, further research is required to enhance our understanding of how prenatal alcohol exposure affects neural plasticity and cognitive functions and to develop effective therapeutic interventions for those impacted. Ultimately, this work aims to advance the comprehension of the consequences of PAE on the brain and cognitive functions.
Collapse
|
7
|
Dyląg KA, Dumnicka P, Kowalska K, Migas-Majoch A, Przybyszewska K, Drożdż D. Increased incidence of renal and urinary tract anomalies among individuals with fetal alcohol spectrum disorders (FASD). Birth Defects Res 2024; 116:e2259. [PMID: 37828651 DOI: 10.1002/bdr2.2259] [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: 10/08/2022] [Revised: 07/24/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) in a spectrum of neurodevelopmental conditions resulting from prenatal alcohol exposure (PAE). Animal models have confirmed the toxic effects of PAE on the kidneys and urinary tract, yet the evidence from human studies is contradictory. The purpose of this study was to establish the incidence of renal and urinary tract anomalies (RUTA), impaired kidney function, and hypertension among patients with FASD. METHODS Children from the FASD Diagnostic Center with FASD diagnosis (FAS, pFAS, or ARND) were offered participation in the study. The control group consisted of patients from the Gastroenterology Department of the same hospital. The patients underwent renal and urinary tract ultrasound examination. The serum creatinine level was also evaluated and the blood pressure was taken twice. Polish OLAF charts were used to determine the percentiles of blood pressure. RESULTS The incidence of kidney and urinary tract defects in the study group was significantly higher than in the control group (OR: 2.64 [1.60-4.34]). The kidney size among FASD patients was significantly lower (73 mm [60-83] vs. 83 mm [70-96]; p < .001) when compared to the control group. No differences were observed in the estimated glomerular filtration rate. In the study group, significantly lower systolic blood, diastolic blood pressure, percentile of systolic pressure, and diastolic pressure were observed. CONCLUSIONS RUTA occurred more frequently among patients with FASD compared to the control group, and decreased kidney size was also demonstrated among patients with FASD. However, impaired kidney function and the risk of hypertension were not observed.
Collapse
Affiliation(s)
- Katarzyna Anna Dyląg
- Department of Patophysiology, Jagiellonian University Medical College, Kraków, małopolskie, Poland
- St. Louis Children Hospital, Kraków, małopolskie, Poland
| | - Paulina Dumnicka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, małopolskie, Poland
| | | | | | | | - Dorota Drożdż
- Department of Paediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, małopolskie, Poland
| |
Collapse
|
8
|
Melder A, Wittmann E, Bulubas L, Dornheim B, Kerber K, Vogelmann U, Campana M, Hubert J, Schmidt V, Heinen F, Padberg F, Landgraf MN. Transcranial magnetic stimulation as a feasible, non-invasive, neuromodulatory intervention in fetal alcohol spectrum disorders. A very first proof of concept. Eur J Paediatr Neurol 2023; 47:131-142. [PMID: 37913649 DOI: 10.1016/j.ejpn.2023.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
INTRODUCTION A neurobiological feature of Fetal Alcohol Spectrum Disorder (FASD) is a global decrease in neuronal connectivity, which leads to significant impairments in everyday functionality. Non-invasive repetitive transcranial magnetic stimulation (rTMS) could potentially positively influence neuronal plasticity but has not yet been studied in FASD. The present trial addresses this gap, making it the first-ever study of rTMS in FASD. MATERIALS AND METHODS The prospective clinical trial was conducted at the LMU University Hospital Munich and enrolled eight FASD participants aged 6-16. Six sessions of 1 Hz-rTMS over the left dorsolateral prefrontal cortex were administered two times a week for three weeks consisting of 1500 pulses at 90 % of resting motor threshold in four trains of 375s. Outcome measures investigated feasibility and treatment response of rTMS on executive functions, attention/impulsivity, social-emotional regulation and quality of life (QoL) via standardized tests and the FASD parents' app. RESULTS Adherence and retention rate were 100 %. Adverse events (AEs) were mild and self-limiting, resulting in a per-session risk of 53.3 %, with local paraesthesia accounting for 54.2 % of the AEs. There were individual relevant but no significant group-level improvements in the investigated functional cerebral domains or participants' QoL. The FASD parents' app showed no significant change in participants' daily functioning or caregivers' QoL. Caregivers' parental stress decreased significantly. CONCLUSION FASD is a very complex disorder that is difficult to treat. In addition, comorbidities as atypical responses to pharmacotherapies are frequent. For this reason, non-invasive, innovative therapies for children with FASD have to be developed. For the first time, rTMS was shown to be safe, tolerable, and acceptable and thus well feasible in paediatric patients with FASD. Further clinical studies with larger samples are needed to identify effective stimulation protocols and to evaluate treatment response.
Collapse
Affiliation(s)
- Anja Melder
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Esther Wittmann
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Lucia Bulubas
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany; Center for Non-invasive Brain Stimulation Munich-Augsburg (CNBS(MA)), Germany
| | - Beate Dornheim
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany; Center for Non-invasive Brain Stimulation Munich-Augsburg (CNBS(MA)), Germany
| | - Katharina Kerber
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Ulrike Vogelmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany; Center for Non-invasive Brain Stimulation Munich-Augsburg (CNBS(MA)), Germany
| | - Mattia Campana
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany; Center for Non-invasive Brain Stimulation Munich-Augsburg (CNBS(MA)), Germany
| | - Jasmin Hubert
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Vivien Schmidt
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Florian Heinen
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Frank Padberg
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany; Center for Non-invasive Brain Stimulation Munich-Augsburg (CNBS(MA)), Germany
| | - Mirjam N Landgraf
- Department of Pediatric Neurology and Developmental Medicine, LMU Center for Development and Children with Medical Complexity, German FASD Competence Center Bavaria, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany.
| |
Collapse
|
9
|
Kukowka A, Brzuchalski B, Kurzawski M, Malinowski D, Białecka MA. ADH1B, ADH1B/C and CYP2E1 Gene Polymorphism and the Risk of Fetal Alcohol Spectrum Disorder. Genes (Basel) 2023; 14:1392. [PMID: 37510297 PMCID: PMC10379323 DOI: 10.3390/genes14071392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Increasing alcohol consumption by women of childbearing age contributes to more frequent cases of fetal alcohol spectrum disorder. The cause of the syndrome is fetal alcohol exposure, particularly what is referred to as high prenatal alcohol exposure. Low metabolic activity of fetal enzymes shifts the burden of ethanol removal to maternal metabolism. One of the factors influencing the pathogenesis of FASD is the genetic background. It can determine the rate of elimination of ethanol, thus increasing or decreasing the time of fetal exposure to ethanol and also decreasing its concentration. Genetic polymorphisms could potentially play a significant role in these processes. In the present study, we considered three polymorphisms of genes implicated in the synthesis of enzymes involved in ethanol metabolism, i.e., ADH1b (rs1229984), ADH1b/c (rs1789891), and CYP2E1 (rs3813867). The studied group consisted of 303 children and 251 mothers. Both mothers' and children's genotypes were considered in our analysis. There were no statistically significant differences between the respective groups of genotypes of the studied polymorphisms. However, the genetic background of FASD is still elusive.
Collapse
Affiliation(s)
- Arnold Kukowka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Bogusław Brzuchalski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland;
| | - Damian Malinowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Monika Anna Białecka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| |
Collapse
|
10
|
Popova S, Charness ME, Burd L, Crawford A, Hoyme HE, Mukherjee RAS, Riley EP, Elliott EJ. Fetal alcohol spectrum disorders. Nat Rev Dis Primers 2023; 9:11. [PMID: 36823161 DOI: 10.1038/s41572-023-00420-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/25/2023]
Abstract
Alcohol readily crosses the placenta and may disrupt fetal development. Harm from prenatal alcohol exposure (PAE) is determined by the dose, pattern, timing and duration of exposure, fetal and maternal genetics, maternal nutrition, concurrent substance use, and epigenetic responses. A safe dose of alcohol use during pregnancy has not been established. PAE can cause fetal alcohol spectrum disorders (FASD), which are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. FASD are a leading preventable cause of birth defects and developmental disability. The prevalence of FASD in 76 countries is >1% and is high in individuals living in out-of-home care or engaged in justice and mental health systems. The social and economic effects of FASD are profound, but the diagnosis is often missed or delayed and receives little public recognition. Future research should be informed by people living with FASD and be guided by cultural context, seek consensus on diagnostic criteria and evidence-based treatments, and describe the pathophysiology and lifelong effects of FASD. Imperatives include reducing stigma, equitable access to services, improved quality of life for people with FASD and FASD prevention in future generations.
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada.
| | - Michael E Charness
- VA Boston Healthcare System, West Roxbury, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Larry Burd
- North Dakota Fetal Alcohol Syndrome Center, Department of Pediatrics, University of North Dakota School of Medicine and Health Sciences, Pediatric Therapy Services, Altru Health System, Grand Forks, ND, USA
| | - Andi Crawford
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - H Eugene Hoyme
- Sanford Children's Genomic Medicine Consortium, Sanford Health, and University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Raja A S Mukherjee
- National UK FASD Clinic, Surrey and Borders Partnership NHS Foundation Trust, Redhill, Surrey, UK
| | - Edward P Riley
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Elizabeth J Elliott
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,New South Wales FASD Assessment Service, CICADA Centre for Care and Intervention for Children and Adolescents affected by Drugs and Alcohol, Sydney Children's Hospitals Network, Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Gasparyan A, Navarro D, Navarrete F, Austrich-Olivares A, Scoma ER, Hambardikar VD, Acosta GB, Solesio ME, Manzanares J. Cannabidiol repairs behavioral and brain disturbances in a model of fetal alcohol spectrum disorder. Pharmacol Res 2023; 188:106655. [PMID: 36642113 DOI: 10.1016/j.phrs.2023.106655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/31/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Fetal alcohol spectrum disorder (FASD) includes neuropsychiatric disturbances related to gestational and lactational ethanol exposure. Available treatments are minimal and do not modulate ethanol-induced damage. Developing animal models simulating FASD is essential for understanding the underlying brain alterations and searching for efficient therapeutic approaches. The main goal of this study was to evaluate the effects of early and chronic cannabidiol (CBD) administration on offspring exposed to an animal model of FASD. Ethanol gavage (3 g/kg/12 h, p.o.) was administered to C57BL/6 J female mice, with a previous history of alcohol consumption, between gestational day 7 and postnatal day 21. On the weaning day, pups were separated by sex, and CBD administration began (30 mg/kg/day, i.p.). After 4-6 weeks of treatment, behavioral and neurobiological changes were analyzed. Mice exposed to the animal model of FASD showed higher anxiogenic and depressive-like behaviors and cognitive impairment that were evaluated through several experimental tests. These behaviors were accompanied by alterations in the gene, cellular and metabolomic targets. CBD administration normalized FASD model-induced emotional and cognitive disturbances, gene expression, and cellular changes with sex-dependent differences. CBD modulates the metabolomic changes detected in the hippocampus and prefrontal cortex. Interestingly, no changes were found in mitochondria or the oxidative status of the cells. These results suggest that the early and repeated administration of CBD modulated the long-lasting behavioral, gene and protein alterations induced by the FASD model, encouraging the possibility of performing clinical trials to evaluate the effects of CBD in children affected with FASD.
Collapse
Affiliation(s)
- Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Amaya Austrich-Olivares
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain
| | - Ernest R Scoma
- Rutgers University, Department of Biology and CCIB, Camden, NJ, USA
| | | | - Gabriela B Acosta
- Instituto de Neurociencias Cognitiva y Traslacional (INCYT), CONICET, INECO, Universidad Favaloro, Ciudad Autónoma de Buenos Aires C1079ABE, Argentina
| | - María E Solesio
- Rutgers University, Department of Biology and CCIB, Camden, NJ, USA
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
12
|
Bauer W, Dylag KA, Lysiak A, Wieczorek-Stawinska W, Pelc M, Szmajda M, Martinek R, Zygarlicki J, Bańdo B, Stomal-Slowinska M, Kawala-Sterniuk A. Initial study on quantitative electroencephalographic analysis of bioelectrical activity of the brain of children with fetal alcohol spectrum disorders (FASD) without epilepsy. Sci Rep 2023; 13:109. [PMID: 36596841 PMCID: PMC9810692 DOI: 10.1038/s41598-022-26590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are spectrum of neurodevelopmental conditions associated with prenatal alcohol exposure. The FASD manifests mostly with facial dysmorphism, prenatal and postnatal growth retardation, and selected birth defects (including central nervous system defects). Unrecognized and untreated FASD leads to severe disability in adulthood. The diagnosis of FASD is based on clinical criteria and neither biomarkers nor imaging tests can be used in order to confirm the diagnosis. The quantitative electroencephalography (QEEG) is a type of EEG analysis, which involves the use of mathematical algorithms, and which has brought new possibilities of EEG signal evaluation, among the other things-the analysis of a specific frequency band. The main objective of this study was to identify characteristic patterns in QEEG among individuals affected with FASD. This study was of a pilot prospective study character with experimental group consisting of patients with newly diagnosed FASD and of the control group consisting of children with gastroenterological issues. The EEG recordings of both groups were obtained, than analyzed using a commercial QEEG module. As a results we were able to establish the dominance of the alpha rhythm over the beta rhythm in FASD-participants compared to those from the control group, mostly in frontal and temporal regions. Second important finding is an increased theta/beta ratio among patients with FASD. These findings are consistent with the current knowledge on the pathological processes resulting from the prenatal alcohol exposure. The obtained results and conclusions were promising, however, further research is necessary (and planned) in order to validate the use of QEEG tools in FASD diagnostics.
Collapse
Affiliation(s)
- Waldemar Bauer
- grid.9922.00000 0000 9174 1488Department of Automatic Control and Robotics, AGH University of Science and Technology, 30-059 Kraków, Poland
| | - Katarzyna Anna Dylag
- St. Louis Children Hospital in Krakow, 30-663 Kraków, Poland ,grid.5522.00000 0001 2162 9631Department of Pathophysiology, Jagiellonian University in Krakow – Collegium Medicum, 31-121 Kraków, Poland
| | - Adam Lysiak
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland
| | | | - Mariusz Pelc
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland ,grid.36316.310000 0001 0806 5472School of Computing and Mathematical Sciences, University of Greenwich, London, SE10 9LS UK
| | - Miroslaw Szmajda
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland
| | - Radek Martinek
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland ,grid.440850.d0000 0000 9643 2828Department of Cybernetics and Biomedical Engineering, VSB—Technical University Ostrava—FEECS, 708 00 Ostrava-Poruba, Czech Republic
| | - Jaroslaw Zygarlicki
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland
| | - Bożena Bańdo
- St. Louis Children Hospital in Krakow, 30-663 Kraków, Poland
| | | | - Aleksandra Kawala-Sterniuk
- grid.440608.e0000 0000 9187 132XFaculty of Electrical Engineering, Automatic Control and Informatics, Opole University of Technology, 45-758 Opole, Poland
| |
Collapse
|
13
|
Fowler CH, Bagdasarov A, Camacho NL, Reuben A, Gaffrey MS. Toxicant exposure and the developing brain: A systematic review of the structural and functional MRI literature. Neurosci Biobehav Rev 2023; 144:105006. [PMID: 36535373 PMCID: PMC9922521 DOI: 10.1016/j.neubiorev.2022.105006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 09/29/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Youth worldwide are regularly exposed to pollutants and chemicals (i.e., toxicants) that may interfere with healthy brain development, and a surge in MRI research has begun to characterize the neurobiological consequences of these exposures. Here, a systematic review following PRISMA guidelines was conducted on developmental MRI studies of toxicants with known or suspected neurobiological impact. Associations were reviewed for 9 toxicant classes, including metals, air pollution, and flame retardants. Of 1264 identified studies, 46 met inclusion criteria. Qualitative synthesis revealed that most studies: (1) investigated air pollutants or metals, (2) assessed exposures prenatally, (3) assessed the brain in late middle childhood, (4) took place in North America or Western Europe, (5) drew samples from existing cohort studies, and (6) have been published since 2017. Given substantial heterogeneity in MRI measures, toxicant measures, and age groups assessed, more research is needed on all toxicants reviewed here. Future studies should also include larger samples, employ personal exposure monitoring, study independent samples in diverse world regions, and assess toxicant mixtures.
Collapse
Affiliation(s)
| | | | | | - Aaron Reuben
- Duke University, 417 Chapel Drive, Durham, NC 27708, USA
| | | |
Collapse
|
14
|
Carpita B, Migli L, Chiarantini I, Battaglini S, Montalbano C, Carmassi C, Cremone IM, Dell’Osso L. Autism Spectrum Disorder and Fetal Alcohol Spectrum Disorder: A Literature Review. Brain Sci 2022; 12:brainsci12060792. [PMID: 35741677 PMCID: PMC9221419 DOI: 10.3390/brainsci12060792] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/07/2022] [Accepted: 06/15/2022] [Indexed: 02/06/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a group of conditions associated with the effects of prenatal alcohol exposure and characterized by somatic and neuropsychological alterations. On the other hand, autism spectrum disorder (ASD) is characterized by a multifaceted neurobehavioral syndrome. Since alcohol can affect every stage of brain development, some authors hypothesized that in utero alcohol exposure might be linked to an increased risk of ASD in subjects with genetic vulnerability. The present review aimed to summarize the available literature on the possible association between FASD and ASD, also focusing on the reported clinical overlaps and on the possible shared pathogenic mechanisms. Studies in this field have stressed similarities and differences between the two conditions, leading to controversial results. The available literature also highlighted that both the disorders are often misdiagnosed or underdiagnosed, stressing the need to broaden the perspective, paying specific attention to milder presentations and sub-syndromic traits.
Collapse
|
15
|
Marguet F, Brosolo M, Friocourt G, Sauvestre F, Marcorelles P, Lesueur C, Marret S, Gonzalez BJ, Laquerrière A. Oligodendrocyte lineage is severely affected in human alcohol-exposed foetuses. Acta Neuropathol Commun 2022; 10:74. [PMID: 35568959 PMCID: PMC9107108 DOI: 10.1186/s40478-022-01378-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/01/2022] [Indexed: 12/12/2022] Open
Abstract
Prenatal alcohol exposure is a major cause of neurobehavioral disabilities. MRI studies in humans have shown that alcohol is associated with white matter microstructural anomalies but these studies focused on myelin abnormalities only after birth. Only one of these studies evaluated oligodendrocyte lineage, but only for a short period during human foetal life. As data are lacking in humans and alcohol is known to impair oligodendrocyte differentiation in rodents, the present study aimed to compare by immunohistochemistry the oligodendrocyte precursor cells expressing PDGFR-α and immature premyelinating/mature oligodendrocytes expressing Olig2 in the ganglionic eminences and the frontal cortex of 14 human foetuses exposed to alcohol from 15 to 37 weeks' gestation with age-matched controls. The human brains used in this study were obtained at the time of foetal autopsies for medical termination of pregnancy, in utero or post-natal early death. Before birth, PDGFR-α expression was strongly increased in the ganglionic eminences and the cortex of all foetuses exposed to alcohol except at the earliest stage. No massive generation of Olig2 immunoreactive cells was identified in the ganglionic eminences until the end of pregnancy and the density of Olig2-positive cells within the cortex was consistently lower in foetuses exposed to alcohol than in controls. These antenatal data from humans provides further evidence of major oligodendrocyte lineage impairment at specific and key stages of brain development upon prenatal alcohol exposure including defective or delayed generation and maturation of oligodendrocyte precursors.
Collapse
Affiliation(s)
- Florent Marguet
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Laboratoire d'Anatomie Pathologique, Pavillon Jacques Delarue, CHU, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 1 Rue de Germont, 76031, Rouen Cedex, France.
| | - Mélanie Brosolo
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Gaëlle Friocourt
- Inserm UMR1078, Université de Bretagne Occidentale, Faculté de Médecine et Des Sciences de la Santé; Etablissement Français du Sang (EFS) Bretagne; Laboratoire de Génétique Moléculaire, CHRU Brest, Hôpital Morvan, Brest, France
| | - Fanny Sauvestre
- Department of Pathology, Bordeaux University Hospital, Bordeaux, France
| | - Pascale Marcorelles
- Pathology Laboratory, Pole Pathologie-Biologie, Centre Hospitalier Universitaire Brest, Brest, France
- Laboratory of Neurosciences of Brest, Faculté de Médecine et des Sciences de la Santé, Brest University, Brest, France
| | - Céline Lesueur
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Stéphane Marret
- Department of Neonatal Paediatrics and Intensive Care, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 76000, Rouen, France
| | - Bruno J Gonzalez
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Laboratoire d'Anatomie Pathologique, Pavillon Jacques Delarue, CHU, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 1 Rue de Germont, 76031, Rouen Cedex, France
| |
Collapse
|
16
|
Sabzali M, Eidi A, Khaksari M, Khastar H. Anti-inflammatory, Antioxidant, and Antiapoptotic Action of Metformin Attenuates Ethanol Neurotoxicity in the Animal Model of Fetal Alcohol Spectrum Disorders. Neurotox Res 2022; 40:605-613. [PMID: 35386022 DOI: 10.1007/s12640-022-00499-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Fetal alcohol exposure has permanent effects on the brain structure, leading to functional deficits in several aspects of behavior, including learning and memory. Alcohol-induced neurocognitive impairment in offsprings is included with activation of oxidative- inflammatory cascade followed with wide apoptotic neurodegeneration in several brain areas, such as the hippocampus. Metformin is the first-line treatment for diabetic patients. It rapidly crosses the blood-brain barrier (BBB) and exerts antioxidant, anti-inflammatory, and neuroprotective effects. In this study, we evaluated the protective effects of metformin on ethanol-related neuroinflammation, as well as neuron apoptosis in the hippocampus of adult male rat in animal model of fetal alcohol spectrum disorders. Treatment with ethanol in milk solution (5.25 and 27.8 g/kg, respectively) was conducted by intragastric intubation at 2-10 days after birth. To examine the antioxidant and anti-inflammatory properties of metformin, an ELISA assay was performed for determining the tumor necrosis factor-α (TNF-α) and antioxidant enzyme concentrations. Immunohistochemical staining was conducted for evaluating the glial fibrillary acidic protein (GFAP) and cleaved caspase-3 expression. Based on the results, metformin caused a significant increase in the superoxide dismutase (SOD) (P < 0.05) and glutathione peroxidase (GSH-Px) (P < 0.01) activities. On the other hand, it reduced the concentrations of TNF-α and malondialdehyde, compared to the ethanol group (P < 0.01). In the metformin group, there was a reduction in cell apoptosis in the hippocampus, as well as GFAP-positive cells (P < 0.01). Overall, apoptotic signaling, regulated by the oxidative inflammatory cascade, can be suppressed by metformin in adult brain rats following animal model of fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Maryam Sabzali
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Hossein Khastar
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
17
|
Trajectories of prenatal alcohol exposure and behavioral outcomes: Findings from a community-based sample. Drug Alcohol Depend 2022; 233:109351. [PMID: 35228080 PMCID: PMC9310559 DOI: 10.1016/j.drugalcdep.2022.109351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To characterize patterns of prenatal alcohol exposure (PAE), and determine whether PAE trajectories were associated with behavior from a community-based sample of first-grade children. METHODS Using data collected as part of the Collaboration of Fetal Alcohol Spectrum Disorders Prevalence study (n = 1663), we performed longitudinal cluster analysis on prenatal alcohol use reported for four time points around conception and pregnancy. From the sample, 638 respondents reported any alcohol use in pregnancy and were included in trajectories for average daily and maximum drinks per drinking day (max DDD). We then estimated the association with behavioral problems measured by the Child Behavior Checklist (CBCL) and Teacher Report Form (TRF) with multivariable linear regression. The reference group had 1025 children with no reported PAE. RESULTS Five trajectories were selected to describe max DDD patterns: very low/discontinuing (n = 186), low/discontinuing (n = 111), very low/continuing (n = 47), med/high (n = 245), and high (n = 49). Six trajectories best described average daily alcohol use: very low/discontinuing (n = 378), very low/continuing (n = 98), low/continuing (n = 56), low/discontinuing (n = 37), medium/high (n = 35), and high (n = 31). When assessing max DDD trajectories for both the CBCL and TRF, individuals with PAE in the two highest trajectories and the very low/continuing trajectory had more behavioral problems relative to children with no PAE, although confidence intervals for most estimates included the null. PAE modeled as average drinks per day did not predict behavior in any consistent pattern. CONCLUSIONS In this community-based sample, select PAE trajectories were associated with behavior, even at relatively low levels of PAE that continued later in gestation.
Collapse
|
18
|
Gillis RF, Palmour RM. mRNA expression analysis of the hippocampus in a vervet monkey model of fetal alcohol spectrum disorder. J Neurodev Disord 2022; 14:21. [PMID: 35305552 PMCID: PMC8934503 DOI: 10.1186/s11689-022-09427-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 02/10/2022] [Indexed: 11/12/2022] Open
Abstract
Background Fetal alcohol spectrum disorders (FASD) are common, yet preventable developmental disorders that stem from prenatal exposure to alcohol. This exposure leads to a wide array of behavioural and physical problems with a complex and poorly defined biological basis. Molecular investigations to date predominantly use rodent animal models, but because of genetic, developmental and social behavioral similarity, primate models are more relevant. We previously reported reduced cortical and hippocampal neuron levels in an Old World monkey (Chlorocebus sabaeus) model with ethanol exposure targeted to the period of rapid synaptogenesis and report here an initial molecular study of this model. The goal of this study was to evaluate mRNA expression of the hippocampus at two different behavioural stages (5 months, 2 years) corresponding to human infancy and early childhood. Methods Offspring of alcohol-preferring or control dams drank a maximum of 3.5 g ethanol per kg body weight or calorically matched sucrose solution 4 days per week during the last 2 months of gestation. Total mRNA expression was measured with the Affymetrix GeneChip Rhesus Macaque Genome Array in a 2 × 2 study design that interrogated two independent variables, age at sacrifice, and alcohol consumption during gestation. Results and discussion Statistical analysis identified a preferential downregulation of expression when interrogating the factor ‘alcohol’ with a balanced effect of upregulation vs. downregulation for the independent variable ‘age’. Functional exploration of both independent variables shows that the alcohol consumption factor generates broad functional annotation clusters that likely implicate a role for epigenetics in the observed differential expression, while the variable age reliably produced functional annotation clusters predominantly related to development. Furthermore, our data reveals a novel connection between EFNB1 and the FASDs; this is highly plausible both due to the role of EFNB1 in neuronal development as well as its central role in craniofrontal nasal syndrome (CFNS). Fold changes for key genes were subsequently confirmed via qRT-PCR. Conclusion Prenatal alcohol exposure leads to global downregulation in mRNA expression. The cellular interference model of EFNB1 provides a potential clue regarding how genetically susceptible individuals may develop the phenotypic triad generally associated with classic fetal alcohol syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-022-09427-z.
Collapse
|
19
|
Harvey DC, De Zoysa P, Toubat O, Choi J, Kishore J, Tsukamoto H, Kumar SR. Concomitant genetic defects potentiate the adverse impact of prenatal alcohol exposure on cardiac outflow tract maturation. Birth Defects Res 2022; 114:105-115. [PMID: 34859965 PMCID: PMC10033225 DOI: 10.1002/bdr2.1968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is associated with an increased incidence of congenital heart defects (CHD), in particular outflow tract (OFT) defects. However, the variability in the incidence of CHD following PAE has not been fully explored. We hypothesize that a concomitant, relevant genetic defect would potentiate the adverse effect of PAE and partially explain the variability of PAE-induced CHD incidence. METHODS The OFT is formed by the second heart field (SHF). Our PAE model consisted of two intraperitoneal injections (3 g/kg, separated by 6 hr) of 30% ethanol on E6.5 during SHF specification. The impact of genetic defects was studied by SHF-specific loss of Delta-like ligand 4 (Dll4), fibroblast growth factor 8 (Fgf8) and Islet1. RESULTS Acute PAE alone significantly increased CHD incidence (4% vs. 26%, p = .015) with a particular increase in OFT alignment defects, viz., double outlet right ventricle (0 vs. 9%, p = .02). In embryos with a SHF genetic defect, acute PAE significantly increased CHD incidence (14 vs. 63%, p < .001), including double outlet right ventricle (6 vs. 50%, p < .001) compared to controls. PAE (p = .01) and heterozygous loss of Dll4 (p = .04) were found to independently contribute to CHD incidence, while neither Islet1 nor Fgf8 defects were found to be significant. CONCLUSIONS Our model recapitulates the increased incidence of OFT alignment defects seen in the clinic due to PAE. The presence of a concomitant SHF genetic mutation increases the incidence of PAE-related OFT defects. An apparent synergistic interaction between PAE and the loss of DLL4-mediated Notch signaling in OFT alignment requires further analysis.
Collapse
Affiliation(s)
- Drayton C Harvey
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Prashan De Zoysa
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Omar Toubat
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Jongkyu Choi
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Jahnavi Kishore
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Hidekazu Tsukamoto
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA
- Greater Los Angeles VA Healthcare System, Los Angeles, California, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| |
Collapse
|
20
|
Dyląg KA, Wieczorek W, Bauer W, Walecki P, Bando B, Martinek R, Kawala-Sterniuk A. Pilot Study on Analysis of Electroencephalography Signals from Children with FASD with the Implementation of Naive Bayesian Classifiers. SENSORS (BASEL, SWITZERLAND) 2021; 22:103. [PMID: 35009650 PMCID: PMC8747358 DOI: 10.3390/s22010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
In this paper Naive Bayesian classifiers were applied for the purpose of differentiation between the EEG signals recorded from children with Fetal Alcohol Syndrome Disorders (FASD) and healthy ones. This work also provides a brief introduction to the FASD itself, explaining the social, economic and genetic reasons for the FASD occurrence. The obtained results were good and promising and indicate that EEG recordings can be a helpful tool for potential diagnostics of FASDs children affected with it, in particular those with invisible physical signs of these spectrum disorders.
Collapse
Affiliation(s)
- Katarzyna Anna Dyląg
- St. Louis Children Hospital, 31-503 Krakow, Poland; (K.A.D.); (B.B.)
- Department of Pathophysiology, Jagiellonian University in Krakow—Collegium Medicum, 31-121 Krakow, Poland
| | - Wiktoria Wieczorek
- Department of Bioinformatics and Telemedicine, Jagiellonian University in Krakow—Collegium Medicum, 30-688 Krakow, Poland; (W.W.); (P.W.)
| | - Waldemar Bauer
- Department of Automatic Control and Robotics, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Piotr Walecki
- Department of Bioinformatics and Telemedicine, Jagiellonian University in Krakow—Collegium Medicum, 30-688 Krakow, Poland; (W.W.); (P.W.)
| | - Bozena Bando
- St. Louis Children Hospital, 31-503 Krakow, Poland; (K.A.D.); (B.B.)
| | - Radek Martinek
- Department of Cybernetics and Biomedical Engineering, VSB—Technical University Ostrava—FEECS, 708 00 Ostrava-Poruba, Czech Republic;
| | | |
Collapse
|
21
|
DiBattista A, Ogrel S, MacKenzie AE, Chakraborty P. Quantitation of phosphatidylethanols in dried blood spots to determine rates of prenatal alcohol exposure in Ontario. Alcohol Clin Exp Res 2021; 46:243-251. [PMID: 34939205 DOI: 10.1111/acer.14766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/04/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Estimating rates of prenatal alcohol exposure (PAE) in a population is necessary to ensure that proper medical and social supports and interventions are in place. This study sought to estimate PAE in Ontario, Canada by quantifying phosphatidylethanol (PEth) homologues in over 2000 residual neonatal dried blood spots (DBS). METHODS A random selection of 2011 residual DBS collected over a 1-week time period were anonymized and extracted. A targeted liquid chromatography-mass spectrometry method was used to quantify 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanol (PEth (16:0/18:1) or POPEth), the clinically accepted biomarker, and six additional PEth homologues. A POPEth level above the United States Drug Testing Laboratories (USDTL) cutoff up to 4 weeks predelivery was indicative of PAE. All PEth homologues were correlated to one another and logistic regression was used to determine the association between PAE status and infant characteristics. RESULTS The estimated rate of PAE in Ontario, up to the last 4 weeks of gestation, was 15.5% (POPEth >28.5 nM). Most PEth homologues were moderately to strongly correlated to one another. A low birth weight and preterm birth were both associated with PAE, while being small for gestational age had lower odds of PAE. CONCLUSIONS The results of this study suggest that PAE may be more prevalent in Ontario than previous estimates by self-report or meconium testing. These findings support the need to consider the effectiveness of current interventions and the design of new interventions to address this significant public health issue.
Collapse
Affiliation(s)
- Alicia DiBattista
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Svetlana Ogrel
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Alex E MacKenzie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Pranesh Chakraborty
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe recent findings on the clinical presentation, pathogenesis, and management of fetal alcohol spectrum disorders (FASDs). Alcohol causes a range of physical, developmental, and cognitive impairments on the developing fetus. Individuals exposed to alcohol prenatally have a wide variability in dysmorphic and neurologic features. Hence, a greater understanding of the mechanisms through which alcohol induces defects in the developing fetus is imperative in developing therapies that prevent alcohol-induced effects. RECENT FINDINGS Current research has focused on leveraging technology to developing tools that can aid in the diagnostic process, defining patterns of neurocognition and neuroimaging specific to FASD, developing neurobehavioral and pharmacologic interventions, and expanding access to care. SUMMARY FASDs are a common cause of neurodevelopmental impairment in school-age children, and their recognition is essential to provide early interventions in order to optimize the outcome for these individuals when they reach adulthood. Although previously thought to be the result of irreversible neurologic injury from prenatal alcohol exposure, recent evidence points to the benefits of applying principles regarding neuroplasticity in improving the lives for patients and their families.
Collapse
Affiliation(s)
- Diego A Gomez
- Mayo Clinic Alix School of Medicine, Phoenix, Arizona
| | - Omar A Abdul-Rahman
- Department of Genetic Medicine, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
23
|
Hen-Herbst L, Tenenbaum A, Senecky Y, Berger A. Pregnant women's alcohol consumption and knowledge about its risks: An Israeli survey. Drug Alcohol Depend 2021; 228:109023. [PMID: 34521056 DOI: 10.1016/j.drugalcdep.2021.109023] [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: 05/03/2021] [Revised: 07/10/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Alcohol consumption is found in a significant proportion of women during their pregnancies. The only study on the prevalence of alcohol consumption during pregnancy in Israel was conducted over a decade ago. Thus, our study aimed to assess alcohol consumption before and during pregnancy, associations with demographic characteristics, knowledge of possible risks of prenatal alcohol exposure, and relations among such knowledge, sociodemographic characteristics, and drinking habits. METHODS A convenience sample of 802 pregnant Israeli women completed an anonymous online questionnaire regarding their alcohol consumption during pregnancy, recommendations received, and knowledge of possible risks. RESULTS Of the sample, 539 (67.2 %) women self-reported drinking alcohol in the 2 months prior to learning they were pregnant, and 96 (12 %) during their pregnancy. Twice as many (28.1 %) reported knowing other women who had consumed alcohol during pregnancy. Women with higher education, in their first pregnancies, ethnically Jewish, and secular reported the highest pre-pregnancy rates of alcohol consumption. About 40 % reported receiving no education about the dangers of alcohol consumption during pregnancy. CONCLUSIONS A concerning percentage of pregnant women in Israel acknowledge drinking alcohol near and after conception. Although most participants reported discontinuing use after realizing they were pregnant, a worrying percentage continued consumption with little knowledge of the dangers. Actual rates may be higher. Information about risks of prenatal alcohol exposure is not widely disseminated, emphasizing the need to increase public awareness.
Collapse
Affiliation(s)
- Liat Hen-Herbst
- Department of Occupational Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel; Department of Occupational Therapy, Faculty of Social Welfare & Health Sciences, University of Haifa, Mount Carmel, Haifa, Israel.
| | - Ariel Tenenbaum
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah Medical Center and the Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Yehuda Senecky
- Department of Pediatric Neurology and Child Development, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Andrea Berger
- Department of Psychology and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
24
|
Licheri V, Chandrasekaran J, Bird CW, Valenzuela CF, Brigman JL. Sex-specific effect of prenatal alcohol exposure on N-methyl-D-aspartate receptor function in orbitofrontal cortex pyramidal neurons of mice. Alcohol Clin Exp Res 2021; 45:1994-2005. [PMID: 34523139 PMCID: PMC8602746 DOI: 10.1111/acer.14697] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/07/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alcohol consumption during pregnancy can produce behavioral and cognitive deficits that persist into adulthood. These include impairments in executive functions, learning, planning, and cognitive flexibility. We have previously shown that moderate prenatal alcohol exposure (PAE) significantly impairs reversal learning, a measure of flexibility mediated across species by different brain areas that include the orbital frontal cortex (OFC). Reversal learning is likewise impaired by genetic or pharmacological inactivation of GluN2B subunit-containing N-methyl-D-aspartate receptors (NMDARs). In the current study, we tested the hypothesis that moderate PAE persistently alters the number and function of GluN2B subunit-containing NMDARs in OFC pyramidal neurons of adult mice. METHODS We used a rodent model of fetal alcohol spectrum disorders and left offspring undisturbed until adulthood. Using whole-cell, patch-clamp recordings, we assessed NMDAR function in slices from 90- to 100-day-old male and female PAE and control mice. Pharmacologically isolated NMDA receptor-mediated evoked excitatory postsynaptic currents (NMDA-eEPSCs) were recorded in the absence and presence of the GluN2B antagonist, Ro25-6981(1 µM). In a subset of littermates, we evaluated the level of GluN2B protein expression in the synaptic fraction using Western blotting technique. RESULTS Our results indicate that PAE females show significantly larger (~23%) NMDA-eEPSC amplitudes than controls, while PAE induced a significant decrease (~17%) in NMDA-eEPSC current density of pyramidal neurons recorded in slices from male mice. NMDA-eEPSC decay time was not affected in PAE-exposed mice from either sex. The contribution of GluN2B subunit-containing NMDARs to the eEPSCs was not significantly altered by PAE. Moreover, there were no significant changes in protein expression in the synaptic fraction of either PAE males or females. CONCLUSIONS These findings suggest that low-to-moderate PAE modulates NMDAR function in pyramidal neurons in a sex-specific manner, although we did not find evidence that the effect is mediated by dysfunction of synaptic GluN2B subunit-containing NMDARs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico
School of Medicine, Albuquerque NM, USA
| | | | - Clark W. Bird
- Department of Neurosciences, University of New Mexico
School of Medicine, Albuquerque NM, USA
| | - C. Fernando Valenzuela
- Department of Neurosciences, University of New Mexico
School of Medicine, Albuquerque NM, USA
- New Mexico Alcohol Research Center, UNM Health Sciences
Center, Albuquerque NM, USA
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico
School of Medicine, Albuquerque NM, USA
- New Mexico Alcohol Research Center, UNM Health Sciences
Center, Albuquerque NM, USA
| |
Collapse
|
25
|
Draghici D, Barr K, Hardy DB, Allman BL, Willmore KE. Effects of advanced maternal age and acute prenatal alcohol exposure on mouse offspring growth and craniofacial phenotype. Alcohol Clin Exp Res 2021; 45:1383-1397. [PMID: 33960427 DOI: 10.1111/acer.14631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) can result in developmental defects that include growth restriction, craniofacial anomalies, and cognitive behavioral deficits, though the presence and severity of these adverse outcomes can vary dramatically among exposed individuals. Preclinical animal models have demonstrated that the dose and timing of PAE account for much, but not all, of this phenotypic variation, suggesting that additional factors mitigate the effects of PAE. Here, we used a mouse model to investigate whether maternal age modulates the effects of PAE on the severity and variation in offspring growth and craniofacial outcomes. METHODS Nulliparous C57BL/6N dams received either an intraperitoneal injection of ethanol (EtOH) or vehicle solution on gestational day 7.5. Dams were divided into four groups: (1) EtOH-treated young dams (6 to 10 weeks); (2) control young dams; (3) EtOH-treated old dams (6 to 7 months); and (4) old control dams. Neonate offspring growth restriction was measured through body mass and organ-to-body mass ratios, while skeletal craniofacial features were imaged using micro-CT and analyzed for size, shape, and variation. RESULTS PAE and advanced maternal age each increased the risk of low birthweight and growth restriction in offspring, but these factors in combination changed the nature of the growth restriction. Similarly, both PAE and advanced maternal age individually caused changes to craniofacial morphology such as smaller skull size, dysmorphic skull shape, and greater skull shape variation and asymmetry. Interestingly, while the combination of PAE and advanced maternal age did not affect mean skull shape or size, it significantly increased the variation and asymmetry of those measures. CONCLUSION Our results indicate that maternal age modulates the effects of PAE, but that the effects of this combination on offspring outcomes are more complex than simply scaling the effects of either factor.
Collapse
Affiliation(s)
- Diana Draghici
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Daniel B Hardy
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Department of Obstetrics and Gynecology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, London, ON, Canada
| | - Brian L Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Katherine E Willmore
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, London, ON, Canada
| |
Collapse
|
26
|
Dylag KA, Bando B, Baran Z, Dumnicka P, Kowalska K, Kulaga P, Przybyszewska K, Radlinski J, Roozen S, Curfs L. Sleep problems among children with Fetal Alcohol Spectrum Disorders (FASD)- an explorative study. Ital J Pediatr 2021; 47:113. [PMID: 34001186 PMCID: PMC8127330 DOI: 10.1186/s13052-021-01056-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/26/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) is a group of conditions resulting from prenatal alcohol exposure (PAE). Patients with FASD experience a variety of neuropsychological symptoms resulting from central nervous system impairment. Little is known about sleep disorders associated with PAE. The objective of this study was to investigate sleep problems related to FASD. METHODS Forty patients (median age 8 years (6; 11)) diagnosed with FASD and forty typically developing children (median age 10 years (8; 13)) were recruited for the 1st phase of the study. In the 1st phase, the screening of sleep problems was performed with Child Sleep Habit Questionnaire (CSHQ) filled in by a caregiver. Those of the FASD group who scored above 41 points were qualified to the 2nd phase of the study and had an in-lab attended polysomnography (PSG) performed. The measurements consisted of electroencephalogram, electrooculograms, chin and tibial electromyogram, electrocardiogram, ventilatory monitoring, breathing effort, pulse oximetry, snoring and body position. Their results were compared to PSG laboratory reference data. RESULTS The number of participants with sleep disturbances was markedly higher in the FASD group as compared to typically developing children (55% vs. 20%). The age-adjusted odds ratio for a positive result in CSHQ was 4.31 (95% CI: 1.54-12.11; p = 0.005) for FASD patients as compared to the control group. Significant differences between the FASD as compared to the typically developing children were observed in the following subscales: sleep onset delay, night wakings, parasomnias, sleep disordered breathing, and daytime sleepiness. Children from the FASD group who underwent PSG experienced more arousals during the sleep as compared with the PSG laboratory reference data. The respiratory indices in FASD group appear higher than previously published data from typically developing children. CONCLUSION The results support the clinical observation that sleep disorders appear to be an important health problem in individuals with FASD. In particular distorted sleep architecture and apneic/hypopneic events need further attention.
Collapse
Affiliation(s)
| | - Bożena Bando
- St. Louis Children Hospital, Strzelecka 2, 31-503, Kraków, Poland
| | - Zbigniew Baran
- National Research Institute for Tuberculosis and Lung Diseases, Rabka Branch, Prof. Jana Rudnika 3B, 34-700, Rabka-Zdrój, Poland
| | - Paulina Dumnicka
- Jagiellonian University Medical College, Department of Medical Diagnostics, Medyczna 9, 30-688, Kraków, Poland
| | | | - Paulina Kulaga
- St. Louis Children Hospital, Strzelecka 2, 31-503, Kraków, Poland
| | | | - Jakub Radlinski
- National Research Institute for Tuberculosis and Lung Diseases, Rabka Branch, Prof. Jana Rudnika 3B, 34-700, Rabka-Zdrój, Poland
| | - Sylvia Roozen
- Governor Kremers Centre, Maastricht University Medical Centre, PO Box 616 6200, MD, Maastricht, The Netherlands
| | - Leopold Curfs
- Governor Kremers Centre, Maastricht University Medical Centre, PO Box 616 6200, MD, Maastricht, The Netherlands
| |
Collapse
|
27
|
Fischer M, Chander P, Kang H, Mellios N, Weick JP. Transcriptomic changes due to early, chronic intermittent alcohol exposure during forebrain development implicate WNT signaling, cell-type specification, and cortical regionalization as primary determinants of fetal alcohol syndrome. Alcohol Clin Exp Res 2021; 45:979-995. [PMID: 33682149 PMCID: PMC8643076 DOI: 10.1111/acer.14590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 01/30/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fetal alcohol syndrome (FAS) due to gestational alcohol exposure represents one of the most common causes of nonheritable lifelong disability worldwide. In vitro and in vivo models have successfully recapitulated multiple facets of the disorder, including morphological and behavioral deficits, but far less is understood regarding the molecular and genetic mechanisms underlying FAS. METHODS In this study, we utilized an in vitro human pluripotent stem cell-based (hPSC) model of corticogenesis to probe the effects of early, chronic intermittent alcohol exposure on the transcriptome of first trimester-equivalent cortical neurons. RESULTS We used RNA sequencing of developing hPSC-derived neurons treated for 50 days with 50 mM ethanol and identified a relatively small number of biological pathways significantly altered by alcohol exposure. These included cell-type specification, axon guidance, synaptic function, and regional patterning, with a notable upregulation of WNT signaling-associated transcripts observed in alcohol-exposed cultures relative to alcohol-naïve controls. Importantly, this effect paralleled a shift in gene expression of transcripts associated with regional patterning, such that caudal forebrain-related transcripts were upregulated at the expense of more anterior ones. Results from H9 embryonic stem cells were largely replicated in an induced pluripotent stem cell line (IMR90-4), indicating that these patterning alterations are not cell line-specific. CONCLUSIONS We found that a major effect of chronic intermittent alcohol on the developing cerebral cortex is an overall imbalance in regionalization, with enrichment of gene expression related to the production of posterodorsal progenitors and a diminution of anteroventral progenitors. This finding parallels behavioral and morphological phenotypes observed in animal models of high-dose prenatal alcohol exposure, as well as patients with FAS.
Collapse
Affiliation(s)
- Máté Fischer
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, NM, USA
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, NM, USA
| | - Huining Kang
- Department of Internal Medicine, University of New Mexico HSC, Albuquerque, NM, USA
| | - Nikolaos Mellios
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, NM, USA.,Autophagy Inflammation and Metabolism (AIM) Center, University of New Mexico HSC, Albuquerque, NM, USA
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, NM, USA.,Center for Brain Recovery and Repair, University of New Mexico HSC, Albuquerque, NM, USA.,New Mexico Alcohol Research Center, University of New Mexico HSC, Albuquerque, NM, USA
| |
Collapse
|
28
|
de Leeuw VC, Pennings JLA, Hessel EVS, Piersma AH. Exploring the biological domain of the neural embryonic stem cell test (ESTn): Morphogenetic regulators, Hox genes and cell types, and their usefulness as biomarkers for embryotoxicity screening. Toxicology 2021; 454:152735. [PMID: 33636252 DOI: 10.1016/j.tox.2021.152735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/25/2021] [Accepted: 02/20/2021] [Indexed: 11/25/2022]
Abstract
Animal-free assessment of compound-induced developmental neurotoxicity will most likely be based on batteries of multiple in vitro tests. The optimal battery is built by combining tests with complementary biological domains that together ideally cover all relevant toxicity pathways. Thus, biological domain definition, i.e. which biological processes and cell types are represented, is an important assay characteristic for determining the place of assays in testing strategies. The murine neural embryonic stem cell test (ESTn) is employed to predict the developmental neurotoxicity of compounds. The aim of this study was to explore the biological domain of ESTn according to three groups of biomarker genes of early (neuro)development: morphogenetic regulators, Hox genes and cell type markers for the ectodermal and neural lineages. These biomarker groups were selected based on their crucial regulatory role in (neuro)development. Analysis of these genes in a series of previously generated whole transcriptome datasets of ESTn showed that at day 7 in culture cell differentiation resembled hindbrain/branchial/thoracic development between E6.5-E12.5 in vivo, with subsequent development into a mixed cell culture containing different neural subtypes, astrocytes and oligodendrocytes by day 13. In addition, the selected biomarkers showed common and distinct responses to compound exposure. Monitoring the biological domain of ESTn through gene expression patterns of morphogenetic regulators, Hox genes and cell type markers proved instrumental in providing mechanistic understanding of compound effects on neural differentiation in ESTn, and can aid in positioning of the test in a battery of complementary in vitro tests in integrated approaches to testing and assessment.
Collapse
Affiliation(s)
- Victoria C de Leeuw
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ellen V S Hessel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
29
|
Mathews E, Dewees K, Diaz D, Favero C. White matter abnormalities in fetal alcohol spectrum disorders: Focus on axon growth and guidance. Exp Biol Med (Maywood) 2021; 246:812-821. [PMID: 33423552 DOI: 10.1177/1535370220980398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fetal Alcohol Spectrum Disorders (FASDs) describe a range of deficits, affecting physical, mental, cognitive, and behavioral function, arising from prenatal alcohol exposure. FASD causes widespread white matter abnormalities, with significant alterations of tracts in the cerebral cortex, cerebellum, and hippocampus. These brain regions present with white-matter volume reductions, particularly at the midline. Neural pathways herein are guided primarily by three guidance cue families: Semaphorin/Neuropilin, Netrin/DCC, and Slit/Robo. These guidance cue/receptor pairs attract and repulse axons and ensure that they reach the proper target to make functional connections. In several cases, these signals cooperate with each other and/or additional molecular partners. Effects of alcohol on guidance cue mechanisms and their associated effectors include inhibition of growth cone response to repellant cues as well as changes in gene expression. Relevant to the corpus callosum, specifically, developmental alcohol exposure alters GABAergic and glutamatergic cell populations and glial cells that serve as guidepost cells for callosal axons. In many cases, deficits seen in FASD mirror aberrancies in guidance cue/receptor signaling. We present evidence for the need for further study on how prenatal alcohol exposure affects the formation of neural connections which may underlie disrupted functional connectivity in FASD.
Collapse
Affiliation(s)
- Erin Mathews
- Biology Department, Ursinus College, Collegeville, PA 19426-1000, USA
| | - Kevyn Dewees
- Biology Department, Ursinus College, Collegeville, PA 19426-1000, USA
| | - Deborah Diaz
- Biology Department, Ursinus College, Collegeville, PA 19426-1000, USA
| | - Carlita Favero
- Biology Department, Ursinus College, Collegeville, PA 19426-1000, USA
| |
Collapse
|
30
|
Díaz-Miranda E, Nadal R, Armario A, Labad J. Prenatal Alcohol Exposure and Hypothalamic-Pituitary-Adrenal Axis Activity of the Offspring in Humans: a Systematic Review. CURRENT ADDICTION REPORTS 2021. [DOI: 10.1007/s40429-020-00349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
31
|
Reid N, Hawkins E, Liu W, Page M, Webster H, Katsikitis M, Shelton D, Wood A, O'Callaghan F, Morrissey S, Shanley D. Yarning about fetal alcohol spectrum disorder: Outcomes of a community-based workshop. RESEARCH IN DEVELOPMENTAL DISABILITIES 2021; 108:103810. [PMID: 33227542 DOI: 10.1016/j.ridd.2020.103810] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/29/2020] [Accepted: 11/05/2020] [Indexed: 06/11/2023]
Abstract
INTRODUCTION There is a lack of neurodevelopmental assessment services in rural and remote locations in Australia that consider fetal alcohol spectrum disorder as a possible outcome. METHODS Eighty-seven participants attended a workshop to support community-based professional development and co-design of a novel assessment approach. Qualitative data collection included video recording of the workshop, and small group discussions, for which a narrative analysis was utilised. Quantitative data collection included self-report questionnaires to understand current community practices and three key constructs: practitioner knowledge, attitudes, and intentions for future practice. RESULTS The narrative analysis highlighted the ongoing impacts of colonisation, in terms of intergenerational trauma and alcohol use, experienced in the community today, and the potential high rates of fetal alcohol spectrum disorder. To address these issues, multiple strategies were discussed, including the recognition of First Nations knowledge and expertise and a focus on the next generation and community organisations working collaboratively. The pre-and post-questionnaires demonstrated that practitioners' knowledge and attitudes were enhanced after attending the workshop, however practitioner intentions were not. The lack of significance for the intentions variable may have been due to the small number of available responses for that variable, in comparison to the other two constructs. DISCUSSION The current study identified key learnings from workshop facilitators and participants. The findings call attention to the importance of a co-design approach, where collaboration is vital to support the appropriate adaption of evidence-based practice to suit the local context.
Collapse
Affiliation(s)
- Natasha Reid
- University of Queensland, Child Health Research Centre, Brisbane, Australia
| | - Erinn Hawkins
- Griffith University, School of Applied Psycholgy, Gold Coast, Australia; Menzies Health Institute of Queensland, Gold Coast, Australia
| | - Wei Liu
- Menzies Health Institute of Queensland, Gold Coast, Australia
| | - Marjad Page
- Menzies Health Institute of Queensland, Gold Coast, Australia; Gidgee Healing, Mount Isa, Australia
| | - Heidi Webster
- Sunshine Coast Health Service, Child Development Service, Sunshine Coast, Australia
| | - Mary Katsikitis
- Flinders University, College of Education, Psychology and Social Work, South Australia, Australia
| | - Doug Shelton
- Griffith University, School of Applied Psycholgy, Gold Coast, Australia; Menzies Health Institute of Queensland, Gold Coast, Australia; Gold Coast University Hospital, Women's and Children's Health Services, Gold Coast, Australia
| | - Andrew Wood
- University of the Sunshine Coast, School of Health and Behavioural Sciences, School of Social Sciences, Sunshine Coast, Australia
| | | | - Shirley Morrissey
- Griffith University, School of Applied Psycholgy, Gold Coast, Australia
| | - Dianne Shanley
- Griffith University, School of Applied Psycholgy, Gold Coast, Australia; Menzies Health Institute of Queensland, Gold Coast, Australia
| |
Collapse
|
32
|
Jawaid S, Strainic JP, Kim J, Ford MR, Thrane L, Karunamuni GH, Sheehan MM, Chowdhury A, Gillespie CA, Rollins AM, Jenkins MW, Watanabe M, Ford SM. Glutathione Protects the Developing Heart from Defects and Global DNA Hypomethylation Induced by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2021; 45:69-78. [PMID: 33206417 PMCID: PMC8865806 DOI: 10.1111/acer.14511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is caused by prenatal alcohol exposure (PAE), the intake of ethanol (C2 H5 OH) during pregnancy. Features of FASD cover a range of structural and functional defects including congenital heart defects (CHDs). Folic acid and choline, contributors of methyl groups to one-carbon metabolism (OCM), prevent CHDs in humans. Using our avian model of FASD, we have previously reported that betaine, another methyl donor downstream of choline, prevents CHDs. The CHD preventions are substantial but incomplete. Ethanol causes oxidative stress as well as depleting methyl groups for OCM to support DNA methylation and other epigenetic alterations. To identify more compounds that can safely and effectively prevent CHDs and other effects of PAE, we tested glutathione (GSH), a compound that regulates OCM and is known as a "master antioxidant." METHODS/RESULTS Quail embryos injected with a single dose of ethanol at gastrulation exhibited congenital defects including CHDs similar to those identified in FASD individuals. GSH injected simultaneously with ethanol not only prevented CHDs, but also improved survival and prevented other PAE-induced defects. Assays of hearts at 8 days (HH stage 34) of quail development, when the heart normally has developed 4-chambers, showed that this single dose of PAE reduced global DNA methylation. GSH supplementation concurrent with PAE normalized global DNA methylation levels. The same assays performed on quail hearts at 3 days (HH stage 19-20) of development, showed no difference in global DNA methylation between controls, ethanol-treated, GSH alone, and GSH plus ethanol-treated cohorts. CONCLUSIONS GSH supplementation shows promise to inhibit effects of PAE by improving survival, reducing the incidence of morphological defects including CHDs, and preventing global hypomethylation of DNA in heart tissues.
Collapse
Affiliation(s)
- Safdar Jawaid
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - James P. Strainic
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Jun Kim
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Matthew R. Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Lars Thrane
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Ganga H. Karunamuni
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Megan M. Sheehan
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Amrin Chowdhury
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Brecksville-Broadview Heights High School, Broadview Heights OH 44147
| | - Caitlyn A. Gillespie
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Fisk University, Nashville TN 37208
| | - Andrew M. Rollins
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michael W. Jenkins
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michiko Watanabe
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Stephanie M Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| |
Collapse
|
33
|
Martín-Estal I, Castilla-Cortázar I, Castorena-Torres F. The Placenta as a Target for Alcohol During Pregnancy: The Close Relation with IGFs Signaling Pathway. Rev Physiol Biochem Pharmacol 2021; 180:119-153. [PMID: 34159446 DOI: 10.1007/112_2021_58] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Alcohol is one of the most consumed drugs in the world, even during pregnancy. Its use is a risk factor for developing adverse outcomes, e.g. fetal death, miscarriage, fetal growth restriction, and premature birth, also resulting in fetal alcohol spectrum disorders. Ethanol metabolism induces an oxidative environment that promotes the oxidation of lipids and proteins, triggers DNA damage, and advocates mitochondrial dysfunction, all of them leading to apoptosis and cellular injury. Several organs are altered due to this harmful behavior, the brain being one of the most affected. Throughout pregnancy, the human placenta is one of the most important organs for women's health and fetal development, as it secretes numerous hormones necessary for a suitable intrauterine environment. However, our understanding of the human placenta is very limited and even more restricted is the knowledge of the impact of toxic substances in its development and fetal growth. So, could ethanol consumption during this period have wounding effects in the placenta, compromising proper fetal organ development? Several studies have demonstrated that alcohol impairs various signaling cascades within G protein-coupled receptors and tyrosine kinase receptors, mainly through its action on insulin and insulin-like growth factor 1 (IGF-1) signaling pathway. This last cascade is involved in cell proliferation, migration, and differentiation and in placentation. This review tries to examine the current knowledge and gaps in our existing understanding of the ethanol effects in insulin/IGFs signaling pathway, which can explain the mechanism to elucidate the adverse actions of ethanol in the maternal-fetal interface of mammals.
Collapse
Affiliation(s)
- Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, Mexico
| | | | | |
Collapse
|
34
|
Li P, Li ZH. Tributyltin Induces the Tissue-Specific Stresses in Zebrafish, a Study in Various Tissues of Muscle, Gill and Intestine. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2020; 105:847-852. [PMID: 33211132 DOI: 10.1007/s00128-020-03048-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/09/2020] [Indexed: 06/11/2023]
Abstract
Because the mechanism of tissue-specific toxicity of tributyltin (TBT) in aquatic organisms has not been explained clearly, the aim of this study is to investigate the effect of chronic exposure to TBT on muscle-related energy metabolism, gill-related ATPase enzymatic system and intestine-related digestive enzymes activities in zebrafish. Male zebrafish were exposed to sub-lethal concentrations of TBT (10, 100 and 300 ng/L) for 6 weeks. Multiple biomarkers were measured (such as glucose, lactate, hexokinase, pyruvate kinase, lactate dehydrogenase, ATP content, ATPases, trypsin, lipase and amylase), which reflected more serious physiological stress with increasing TBT concentrations during the experimental period. Through principal component analysis (PCA) and integrated biomarker response (IBR) analysis, the toxic effect of TBT in zebrafish was in a concentration-dependent manner. Shortly, the results of this study can provide new evidence for a comprehensive understanding of the toxic effects of TBT.
Collapse
Affiliation(s)
- Ping Li
- Marine College, Shandong University, Weihai, 264209, Shandong, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, 264209, Shandong, China.
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China.
| |
Collapse
|
35
|
Wynn A, Rotheram-Borus MJ, Davis E, le Roux I, Almirol E, O'Connor M, Tomlinson M. Identifying fetal alcohol spectrum disorder among South African children at aged 1 and 5 years. Drug Alcohol Depend 2020; 217:108266. [PMID: 32956976 PMCID: PMC7736512 DOI: 10.1016/j.drugalcdep.2020.108266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Fetal Alcohol Spectrum Disorders (FASD) are a global health concern. Early intervention mitigates deficits, yet early diagnosis remains challenging. We examined whether children can be screened and meet diagnoses for FASD at 1.5 years compared to 5 years post-birth. METHODS A population cohort of pregnant women in 24 neighborhoods (N = 1258) was recruited and 84.5 %-96 % were reassessed at two weeks post-birth, 0.5 years, 1.5 years, 3 years, and 5 years later. A two-step process was followed to diagnose FASD; first, a paraprofessional screened the children and then a physician evaluated the child. We evaluated FASD symptoms at 1.5 vs. 5 years. We also examined maternal differences in children receiving a positive FASD screening (n = 160) with those who received a negative FASD screening. RESULTS Screening positive for FASD more than doubled from 1.5 years to 5 years (from 6.8 % to 14.8 %). About one quarter of children who screened positive and were evaluated by a physician, were diagnosed as having a FASD. However, half did not complete the 2nd stage screening. Compared to mothers of children with a negative FASD screening, mothers of children with a positive FASD screening were less likely to have a high school education and more likely to have lower incomes, have experienced interpersonal partner violence, and have a depressed mood. Mothers of children who did not follow up for a 2nd stage physician evaluation were more like to live in informal housing compared to those who followed-up (81.3 % vs. 62.5 %, p = 0.014). CONCLUSIONS We found that children can be screened and diagnosed for FASD at 1.5 and 5 years. As FASD characteristics develop over time, repeated screenings are necessary to identify all affected children and launch preventive interventions. Referrals for children to see a physician to confirm diagnosis and link children to care remains a challenge. Integration with the primary healthcare system might mitigate some of those difficulties.
Collapse
Affiliation(s)
- Adriane Wynn
- Division of Infectious Diseases and Global Public Health, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92024, USA
| | - Mary Jane Rotheram-Borus
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California at Los Angeles, 10920 Wilshire Blvd., Suite 350, Los Angeles, CA 90024, USA.
| | - Emily Davis
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California at Los Angeles, 10920 Wilshire Blvd., Suite 350, Los Angeles, CA 90024, USA
| | - Ingrid le Roux
- Philani Child Health and Nutrition Project, Khayelitsha, South Africa
| | - Ellen Almirol
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California at Los Angeles, 10920 Wilshire Blvd., Suite 350, Los Angeles, CA 90024, USA
| | - Mary O'Connor
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California at Los Angeles, 10920 Wilshire Blvd., Suite 350, Los Angeles, CA 90024, USA
| | - Mark Tomlinson
- Institute for Life Course Health Research, Department of Global Health, Stellenbosch University, Cape Town, South Africa; School of Nursing and Midwifery, Queens University, Belfast, UK
| |
Collapse
|
36
|
Arzua T, Yan Y, Jiang C, Logan S, Allison RL, Wells C, Kumar SN, Schäfer R, Bai X. Modeling alcohol-induced neurotoxicity using human induced pluripotent stem cell-derived three-dimensional cerebral organoids. Transl Psychiatry 2020; 10:347. [PMID: 33051447 PMCID: PMC7553959 DOI: 10.1038/s41398-020-01029-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Maternal alcohol exposure during pregnancy can substantially impact the development of the fetus, causing a range of symptoms, known as fetal alcohol spectrum disorders (FASDs), such as cognitive dysfunction and psychiatric disorders, with the pathophysiology and mechanisms largely unknown. Recently developed human cerebral organoids from induced pluripotent stem cells are similar to fetal brains in the aspects of development and structure. These models allow more relevant in vitro systems to be developed for studying FASDs than animal models. Modeling binge drinking using human cerebral organoids, we sought to quantify the downstream toxic effects of alcohol (ethanol) on neural pathology phenotypes and signaling pathways within the organoids. The results revealed that alcohol exposure resulted in unhealthy organoids at cellular, subcellular, bioenergetic metabolism, and gene expression levels. Alcohol induced apoptosis on organoids. The apoptotic effects of alcohol on the organoids depended on the alcohol concentration and varied between cell types. Specifically, neurons were more vulnerable to alcohol-induced apoptosis than astrocytes. The alcohol-treated organoids exhibit ultrastructural changes such as disruption of mitochondria cristae, decreased intensity of mitochondrial matrix, and disorganized cytoskeleton. Alcohol exposure also resulted in mitochondrial dysfunction and metabolic stress in the organoids as evidenced by (1) decreased mitochondrial oxygen consumption rates being linked to basal respiration, ATP production, proton leak, maximal respiration and spare respiratory capacity, and (2) increase of non-mitochondrial respiration in alcohol-treated organoids compared with control groups. Furthermore, we found that alcohol treatment affected the expression of 199 genes out of 17,195 genes analyzed. Bioinformatic analyses showed the association of these dysregulated genes with 37 pathways related to clinically relevant pathologies such as psychiatric disorders, behavior, nervous system development and function, organismal injury and abnormalities, and cellular development. Notably, 187 of these genes are critically involved in neurodevelopment, and/or implicated in nervous system physiology and neurodegeneration. Furthermore, the identified genes are key regulators of multiple pathways linked in networks. This study extends for the first time animal models of binge drinking-related FASDs to a human model, allowing in-depth analyses of neurotoxicity at tissue, cellular, subcellular, metabolism, and gene levels. Hereby, we provide novel insights into alcohol-induced pathologic phenotypes, cell type-specific vulnerability, and affected signaling pathways and molecular networks, that can contribute to a better understanding of the developmental neurotoxic effects of binge drinking during pregnancy.
Collapse
Affiliation(s)
- Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Yasheng Yan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Congshan Jiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Reilly L Allison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Clive Wells
- Department of Microbiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Suresh N Kumar
- Department of Pathology, Children's Research Institute Imaging Core, Neuroscience Imaging Facility, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, 60438, Frankfurt am Main, Germany
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA.
| |
Collapse
|
37
|
Léger C, Dupré N, Laquerrière A, Lecointre M, Dumanoir M, Janin F, Hauchecorne M, Fabre M, Jégou S, Frébourg T, Cleren C, Leroux P, Marcorelles P, Brasse-Lagnel C, Marret S, Marguet F, Gonzalez BJ. In utero alcohol exposure exacerbates endothelial protease activity from pial microvessels and impairs GABA interneuron positioning. Neurobiol Dis 2020; 145:105074. [PMID: 32890773 DOI: 10.1016/j.nbd.2020.105074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
In utero alcohol exposure can induce severe neurodevelopmental disabilities leading to long-term behavioral deficits. Because alcohol induces brain defects, many studies have focused on nervous cells. However, recent reports have shown that alcohol markedly affects cortical angiogenesis in both animal models and infants with fetal alcohol spectrum disorder (FASD). In addition, the vascular system is known to contribute to controlling gamma-aminobutyric acid (GABA)ergic interneuron migration in the developing neocortex. Thus, alcohol-induced vascular dysfunction may contribute to the neurodevelopmental defects in FASD. The present study aimed at investigating the effects of alcohol on endothelial activity of pial microvessels. Ex vivo experiments on cortical slices from mouse neonates revealed that in endothelial cells from pial microvessels acute alcohol exposure inhibits both glutamate-induced calcium mobilization and activities of matrix metalloproteinase-9 (MMP-9) and tissue plasminogen activator (tPA). The inhibitory effect of alcohol on glutamate-induced MMP-9 activity was abrogated in tPA-knockout and Grin1flox/VeCadcre mice suggesting that alcohol interacts through the endothelial NMDAR/tPA/MMP-9 vascular pathway. Contrasting with the effects from acute alcohol exposure, in mouse neonates exposed to alcohol in utero during the last gestational week, glutamate exacerbated both calcium mobilization and endothelial protease activities from pial microvessels. This alcohol-induced vascular dysfunction was associated with strong overexpression of the N-methyl-d-aspartate receptor subunit GluN1 and mispositioning of the Gad67-GFP interneurons that normally populate the superficial cortical layers. By comparing several human control fetuses with a fetus chronically exposed to alcohol revealed that alcohol exposure led to mispositioning of the calretinin-positive interneurons, whose density was decreased in the superficial cortical layers II-III and increased in deepest layers. This study provides the first mechanistic and functional evidence that alcohol impairs glutamate-regulated activity of pial microvessels. Endothelial dysfunction is characterized by altered metalloproteinase activity and interneuron mispositioning, which was also observed in a fetus with fetal alcohol syndrome. These data suggest that alcohol-induced endothelial dysfunction may contribute in ectopic cortical GABAergic interneurons, that has previously been described in infants with FASD.
Collapse
Affiliation(s)
- Cécile Léger
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Nicolas Dupré
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Annie Laquerrière
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Maryline Lecointre
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Marion Dumanoir
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - François Janin
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Michelle Hauchecorne
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Maëlle Fabre
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Sylvie Jégou
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frébourg
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Carine Cleren
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Philippe Leroux
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | | | - Carole Brasse-Lagnel
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Florent Marguet
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Bruno J Gonzalez
- Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France.
| |
Collapse
|
38
|
Buján GE, Serra HA, Molina SJ, Guelman LR. Oxidative Stress-Induced Brain Damage Triggered by Voluntary Ethanol Consumption during Adolescence: A Potential Target for Neuroprotection? Curr Pharm Des 2020; 25:4782-4790. [PMID: 31814553 DOI: 10.2174/1381612825666191209121735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/23/2019] [Indexed: 12/28/2022]
Abstract
Alcohol consumption, in particular ethanol (EtOH), typically begins in human adolescence, often in a "binge like" manner. However, although EtOH abuse has a high prevalence at this stage, the effects of exposure during adolescence have been less explored than prenatal or adult age exposure. Several authors have reported that EtOH intake during specific periods of development might induce brain damage. Although the mechanisms are poorly understood, it has been postulated that oxidative stress may play a role. In fact, some of these studies revealed a decrease in brain antioxidant enzymes' level and/or an increase in reactive oxygen species (ROS) production. Nevertheless, although existing literature shows a number of studies in which ROS were measured in developing animals, fewer reported the measurement of ROS levels after EtOH exposure in adolescence. Importantly, neuroprotective agents aimed to these potential targets may be relevant tools useful to reduce EtOH-induced neurodegeneration, restore cognitive function and improve treatment outcomes for alcohol use disorders (AUDs). The present paper reviews significant evidences about the mechanisms involved in EtOH-induced brain damage, as well as the effect of different potential neuroprotectants that have shown to be able to prevent EtOH-induced oxidative stress. A selective inhibitor of the endocannabinoid anandamide metabolism, a flavonol present in different fruits (quercetin), an antibiotic with known neuroprotective properties (minocycline), a SOD/catalase mimetic, a potent antioxidant and anti-inflammatory molecule (resveratrol), a powerful ROS scavenger (melatonin), an isoquinoline alkaloid (berberine), are some of the therapeutic strategies that could have some clinical relevance in the treatment of AUDs. As most of these works were performed in adult animal models and using EtOH-forced paradigms, the finding of neuroprotective tools that could be effective in adolescent animal models of voluntary EtOH intake should be encouraged.
Collapse
Affiliation(s)
- Gustavo E. Buján
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| | - Hector A. Serra
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina
| | - Sonia J. Molina
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| | - Laura R. Guelman
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| |
Collapse
|
39
|
Gibson S, Nagle C, Paul J, McCarthy L, Muggli E. Influences on drinking choices among Indigenous and non-Indigenous pregnant women in Australia: A qualitative study. PLoS One 2020; 15:e0224719. [PMID: 32352991 PMCID: PMC7192424 DOI: 10.1371/journal.pone.0224719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/13/2020] [Indexed: 12/24/2022] Open
Abstract
Despite women's awareness that drinking alcohol in pregnancy can lead to lifelong disabilities in a child, it appears that an awareness alone does not discourage some pregnant women from drinking. To explore influences on pregnant women's choices around alcohol use, we conducted interviews and group discussions with 14 Indigenous Australian and 14 non-Indigenous pregnant women attending antenatal care in a range of socioeconomic settings. Inductive content analysis identified five main influences on pregnant women's alcohol use: the level and detail of women's understanding of harm; women's information sources on alcohol use in pregnancy; how this information influenced their choices; how women conceptualised their pregnancy; and whether the social and cultural environment supported abstinence. Results provide insight into how Indigenous Australian and non-Indigenous pregnant women understand and conceptualise the harms from drinking alcohol when making drinking choices, including how their social and cultural environments impact their ability to abstain. Strategies for behaviour change need to: correct misinformation about supposed 'safe' timing, quantity and types of alcohol; develop a more accurate perception of Fetal Alcohol Spectrum Disorder; reframe messages about harm to messages about optimising the child's health and cognitive outcomes; and develop a holistic approach encompassing women's social and cultural context.
Collapse
Affiliation(s)
- Sophie Gibson
- Reproductive Epidemiology, Murdoch Children's Research Institute, Parkville, Australia
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, Australia
| | - Cate Nagle
- Centre for Nursing & Midwifery Research, James Cook University, Townsville, Australia
- Townsville Hospital & Health Service, Townsville, Australia
| | - Jean Paul
- Reproductive Epidemiology, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Leisa McCarthy
- Menzies School of Health Research, Alice Springs, Australia
| | - Evelyne Muggli
- Reproductive Epidemiology, Murdoch Children's Research Institute, Parkville, Australia
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, Australia
- Townsville Hospital & Health Service, Townsville, Australia
| |
Collapse
|
40
|
Subramanian L, Calcagnotto ME, Paredes MF. Cortical Malformations: Lessons in Human Brain Development. Front Cell Neurosci 2020; 13:576. [PMID: 32038172 PMCID: PMC6993122 DOI: 10.3389/fncel.2019.00576] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Creating a functional cerebral cortex requires a series of complex and well-coordinated developmental steps. These steps have evolved across species with the emergence of cortical gyrification and coincided with more complex behaviors. The presence of diverse progenitor cells, a protracted timeline for neuronal migration and maturation, and diverse neuronal types are developmental features that have emerged in the gyrated cortex. These factors could explain how the human brain has expanded in size and complexity. However, their complex nature also renders new avenues of vulnerability by providing additional cell types that could contribute to disease and longer time windows that could impact the composition and organization of the cortical circuit. We aim to discuss the unique developmental steps observed in human corticogenesis and propose how disruption of these species-unique processes could lead to malformations of cortical development.
Collapse
Affiliation(s)
- Lakshmi Subramanian
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mercedes F Paredes
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States.,Neuroscience Graduate Division, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
41
|
Fainsod A, Bendelac-Kapon L, Shabtai Y. Fetal Alcohol Spectrum Disorder: Embryogenesis Under Reduced Retinoic Acid Signaling Conditions. Subcell Biochem 2020; 95:197-225. [PMID: 32297301 DOI: 10.1007/978-3-030-42282-0_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a complex set of developmental malformations, neurobehavioral anomalies and mental disabilities induced by exposing human embryos to alcohol during fetal development. Several experimental models and a series of developmental and biochemical approaches have established a strong link between FASD and reduced retinoic acid (RA) signaling. RA signaling is involved in the regulation of numerous developmental decisions from patterning of the anterior-posterior axis, starting at gastrulation, to the differentiation of specific cell types within developing organs, to adult tissue homeostasis. Being such an important regulatory signal during embryonic development, mutations or environmental perturbations that affect the level, timing or location of the RA signal can induce multiple and severe developmental malformations. The evidence connecting human syndromes to reduced RA signaling is presented here and the resulting phenotypes are compared to FASD. Available data suggest that competition between ethanol clearance and RA biosynthesis is a major etiological component in FASD.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel.
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| |
Collapse
|
42
|
Franks AL, Berry KJ, DeFranco DB. Prenatal drug exposure and neurodevelopmental programming of glucocorticoid signalling. J Neuroendocrinol 2020; 32:e12786. [PMID: 31469457 PMCID: PMC6982551 DOI: 10.1111/jne.12786] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Prenatal neurodevelopment is dependent on precise functioning of multiple signalling pathways in the brain, including those mobilised by glucocorticoids (GC) and endocannabinoids (eCBs). Prenatal exposure to drugs of abuse, including opioids, alcohol, cocaine and cannabis, has been shown to not only impact GC signalling, but also alter functioning of the hypothalamic-pituitary-adrenal (HPA) axis. Such exposures can have long-lasting neurobehavioural consequences, including alterations in the stress response in the offspring. Furthermore, cannabis contains cannabinoids that signal via the eCB pathway, which is linked to some components of GC signalling in the adult brain. Given that GCs are frequently used in pregnancy to prevent complications of prematurity, and also that rates of cannabis use in pregnancy are increasing, the likelihood of foetal co-exposure to these compounds is high and may have additional implications for long-term neurodevelopment. Here, we present a discussion of GC signalling and the HPA axis, as well as the effects of prenatal drug exposure on these pathways and the stress response, and we explore the interactions between GC and EC signalling in the developing brain and potential for neurodevelopmental consequences.
Collapse
Affiliation(s)
- Alexis L Franks
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kimberly J Berry
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald B DeFranco
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology and Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Grandjean P, Abdennebi-Najar L, Barouki R, Cranor CF, Etzel RA, Gee D, Heindel JJ, Hougaard KS, Hunt P, Nawrot TS, Prins GS, Ritz B, Soffritti M, Sunyer J, Weihe P. Timescales of developmental toxicity impacting on research and needs for intervention. Basic Clin Pharmacol Toxicol 2019; 125 Suppl 3:70-80. [PMID: 30387920 PMCID: PMC6497561 DOI: 10.1111/bcpt.13162] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/29/2018] [Indexed: 12/28/2022]
Abstract
Much progress has happened in understanding developmental vulnerability to preventable environmental hazards. Along with the improved insight, the perspective has widened, and developmental toxicity now involves latent effects that can result in delayed adverse effects in adults or at old age and additional effects that can be transgenerationally transferred to future generations. Although epidemiology and toxicology to an increasing degree are exploring the adverse effects from developmental exposures in human beings, the improved documentation has resulted in little progress in protection, and few environmental chemicals are currently regulated to protect against developmental toxicity, whether it be neurotoxicity, endocrine disruption or other adverse outcome. The desire to obtain a high degree of certainty and verification of the evidence used for decision-making must be weighed against the costs and necessary duration of research, as well as the long-term costs to human health because of delayed protection of vulnerable early-life stages of human development and, possibly, future generations. Although two-generation toxicology tests may be useful for initial test purposes, other rapidly emerging tools need to be seriously considered from computational chemistry and metabolomics to CLARITY-BPA-type designs, big data and population record linkage approaches that will allow efficient generation of new insight; epigenetic mechanisms may necessitate a set of additional regulatory tests to reveal such effects. As reflected by the Prenatal Programming and Toxicity (PPTOX) VI conference, the current scientific understanding and the timescales involved require an intensified approach to protect against preventable adverse health effects that can harm the next generation and generations to come. While further research is needed, the main emphasis should be on research translation and timely public health intervention to avoid serious, irreversible and perhaps transgenerational harm.
Collapse
Affiliation(s)
- Philippe Grandjean
- Department of Environmental Medicine, University of Southern Denmark, Odense, Denmark
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Robert Barouki
- INSERM UMR-S 1124, Université Paris Descartes, Paris, France
| | - Carl F Cranor
- Department of Philosophy, University of California, Riverside, California
| | - Ruth A Etzel
- Milken Institute, School of Public Health, The George Washington University, Washington, District of Columbia
| | - David Gee
- Institute of Environment, Health and Societies, Brunel University, London, UK
| | - Jerrold J Heindel
- Program in Endocrine Disruption Strategies, Commonweal, Bolinas, California
| | - Karin S Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Patricia Hunt
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Department of Public Health, Leuven University, Leuven, Belgium
| | - Gail S Prins
- Chicago Center for Health and Environment (CACHET), University of Illinois at Chicago, Chicago, Illinois
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California
| | - Morando Soffritti
- Ramazzini Institute, Bologna, Italy
- European Foundation for Cancer Research, Environmental and Occupational Diseases "Ruberti Schileo", Treviso, Italy
| | - Jordi Sunyer
- ISGlobal, Centre for Research in Environmental Epidemiology, Barcelona, Catalonia, Spain
| | - Pal Weihe
- Department of Public Health and Occupational Medicine, Tórshavn, Faroe Islands
| |
Collapse
|
44
|
Bhatia S, Drake DM, Miller L, Wells PG. Oxidative stress and DNA damage in the mechanism of fetal alcohol spectrum disorders. Birth Defects Res 2019; 111:714-748. [PMID: 31033255 DOI: 10.1002/bdr2.1509] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022]
Abstract
This review covers molecular mechanisms involving oxidative stress and DNA damage that may contribute to morphological and functional developmental disorders in animal models resulting from exposure to alcohol (ethanol, EtOH) in utero or in embryo culture. Components covered include: (a) a brief overview of EtOH metabolism and embryopathic mechanisms other than oxidative stress; (b) mechanisms within the embryo and fetal brain by which EtOH increases the formation of reactive oxygen species (ROS); (c) critical embryonic/fetal antioxidative enzymes and substrates that detoxify ROS; (d) mechanisms by which ROS can alter development, including ROS-mediated signal transduction and oxidative DNA damage, the latter of which leads to pathogenic genetic (mutations) and epigenetic changes; (e) pathways of DNA repair that mitigate the pathogenic effects of DNA damage; (f) related indirect mechanisms by which EtOH enhances risk, for example by enhancing the degradation of some DNA repair proteins; and, (g) embryonic/fetal pathways like NRF2 that regulate the levels of many of the above components. Particular attention is paid to studies in which chemical and/or genetic manipulation of the above mechanisms has been shown to alter the ability of EtOH to adversely affect development. Alterations in the above components are also discussed in terms of: (a) individual embryonic and fetal determinants of risk and (b) potential risk biomarkers and mitigating strategies. FASD risk is likely increased in progeny which/who are biochemically predisposed via genetic and/or environmental mechanisms, including enhanced pathways for ROS formation and/or deficient pathways for ROS detoxification or DNA repair.
Collapse
Affiliation(s)
- Shama Bhatia
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Danielle M Drake
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | | | - Peter G Wells
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Tanguy E, Wang Q, Moine H, Vitale N. Phosphatidic Acid: From Pleiotropic Functions to Neuronal Pathology. Front Cell Neurosci 2019; 13:2. [PMID: 30728767 PMCID: PMC6351798 DOI: 10.3389/fncel.2019.00002] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/07/2019] [Indexed: 11/17/2022] Open
Abstract
Among the cellular lipids, phosphatidic acid (PA) is a peculiar one as it is at the same time a key building block of phospholipid synthesis and a major lipid second messenger conveying signaling information. The latter is thought to largely occur through the ability of PA to recruit and/or activate specific proteins in restricted compartments and within those only at defined submembrane areas. Furthermore, with its cone-shaped geometry PA locally changes membrane topology and may thus be a key player in membrane trafficking events, especially in membrane fusion and fission steps, where lipid remodeling is believed to be crucial. These pleiotropic cellular functions of PA, including phospholipid synthesis and homeostasis together with important signaling activity, imply that perturbations of PA metabolism could lead to serious pathological conditions. In this mini-review article, after outlining the main cellular functions of PA, we highlight the different neurological diseases that could, at least in part, be attributed to an alteration in PA synthesis and/or catabolism.
Collapse
Affiliation(s)
- Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR-3212 Centre National de la Recherche Scientifique & Université de Strasbourg, Strasbourg, France
| | - Qili Wang
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR-3212 Centre National de la Recherche Scientifique & Université de Strasbourg, Strasbourg, France
| | - Hervé Moine
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR-3212 Centre National de la Recherche Scientifique & Université de Strasbourg, Strasbourg, France
| |
Collapse
|
46
|
Roozen S, Peters GJY, Kok G, Townend D, Nijhuis J, Koek G, Curfs L. Systematic literature review on which maternal alcohol behaviours are related to fetal alcohol spectrum disorders (FASD). BMJ Open 2018; 8:e022578. [PMID: 30573481 PMCID: PMC6303602 DOI: 10.1136/bmjopen-2018-022578] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Fetal alcohol spectrum disorders (FASD) is a worldwide problem. Maternal alcohol consumption is an important risk factor for FASD. It remains unknown which alcohol consumption patterns most strongly predict FASD. The objective of this study was to identify these. DESIGN Systematic literature review. METHODS We searched in PubMed, PsychINFO, PsycARTICLES, ERIC, CINAHL, Embase and MEDLINE up to August 2018. The query consisted of keywords and their synonyms related to FASD, pregnancy and behaviour. Studies were excluded when not published in English, were reviews or involved non-human subjects. Substantial heterogeneity precluded aggregation or meta-analysis of the data. Instead, data were qualitatively inspected. RESULTS In total, 21 studies were eligible for further data analysis. All studies that measured both maternal alcohol drinking behaviours and FASD reported retrospective data on maternal drinking patterns, employing both continuous and categorical measures and exhibiting substantial heterogeneity in measures of alcohol consumption (eg, timing of exposure, quantification of alcohol measure and definition of a standard drink). Study quality improved over time and appeared higher for studies based on active case ascertainment, especially when conducted in schools and when behaviour was assessed through interviews. CONCLUSIONS We aimed to identify specific maternal drinking behaviour(s) related to FASD. The state of the literature precludes such conclusions. Evidence-based preventive measures necessitate identifying which prenatal alcohol drinking behaviour(s) are most in need of intervention. Therefore, we formulate three recommendations for future research. First, future studies can optimise the value of the collected dataset through specifying measurements and reporting of maternal drinking behaviours and avoiding categorised measures (nominal or ordinal) whenever possible. Second, samples should not be selected based on FASD status, but instead, FASD status as well as maternal alcohol consumption should both be measured in a general population sample. Finally, we provide 10 reporting guidelines for FASD research.
Collapse
Affiliation(s)
- Sylvia Roozen
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Work and Social Psychology, Maastricht University, Maastricht, The Netherlands
| | - Gjalt-Jorn Ygram Peters
- Department of Work and Social Psychology, Maastricht University, Maastricht, The Netherlands
- Faculty of Psychology and Education Science, Open University of The Netherlands, Heerlen, The Netherlands
| | - Gerjo Kok
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Work and Social Psychology, Maastricht University, Maastricht, The Netherlands
| | - David Townend
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Health, Ethics & Society, Maastricht University, Maastricht, The Netherlands
| | - Jan Nijhuis
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Obstretrics and Gynaecology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ger Koek
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leopold Curfs
- Governor Kremers Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|