1
|
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
|
2
|
Fletcher TMD, Mullan B, Finlay-Jones A. Intention to Engage in Alcohol Use during Pregnancy: The Role of Attitudes and Prototypes. Subst Use Misuse 2023; 58:1333-1342. [PMID: 37249357 DOI: 10.1080/10826084.2023.2215292] [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] [Indexed: 05/31/2023]
Abstract
Background: There is no known risk-free level of alcohol use in pregnancy. Despite this, many still believe that occasional drinking is safe. To-date, there is limited evidence of the influences on women's decisions about low to moderate alcohol use in pregnancy. The aim of this study was to explore alcohol use intentions during pregnancy, using variables from the theory of planned behavior, the prototype/willingness model and personality variables. The study also investigated whether priming participants with exposure to prototypes describing different alcohol use behaviors had an impact on intentions.Methods: Participants, 746 women aged 20 to 45 years, were randomized to be prompted to think of one of two different "types" of behaviors, i.e., small level of alcohol use in pregnancy and ambiguous level of alcohol use in pregnancy. They then completed measures of theoretical variables, impulsivity, venturesomeness, and self-efficacy. Participants then answered whether they intended to use alcohol during a future pregnancy.Results: Over half of the variance in intentions to consume alcohol while pregnant were predicted by the final model (R2= .527, F (1, 438) = 13.201, p < .001). Positive attitudes toward alcohol use in pregnancy, from the theory of planned behavior, were the most significant predictor of intentions and intentions did not differ between groups according to prototype exposure.Conclusions: Future research should aim to explore the efficacy of interventions to reduce low to moderate alcohol use in pregnancy that utilize both the theory of planned behavior and prototype/willingness model to target determinants of intentions.
Collapse
Affiliation(s)
- Tess M D Fletcher
- Enable Institute, Curtin University, Perth, Australia
- FASD Research Australia Centre for Research Excellence, Telethon Kids Institute, Perth, Australia
| | | | - Amy Finlay-Jones
- FASD Research Australia Centre for Research Excellence, Telethon Kids Institute, Perth, Australia
| |
Collapse
|
3
|
Rodríguez-Sastre N, Shapiro N, Hawkins DY, Lion AT, Peyreau M, Correa AE, Dionne K, Bradham CA. Ethanol exposure perturbs sea urchin development and disrupts developmental timing. Dev Biol 2023; 493:89-102. [PMID: 36368523 DOI: 10.1016/j.ydbio.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Ethanol is a known vertebrate teratogen that causes craniofacial defects as a component of fetal alcohol syndrome (FAS). Our results show that sea urchin embryos treated with ethanol similarly show broad skeletal patterning defects, potentially analogous to the defects associated with FAS. The sea urchin larval skeleton is a simple patterning system that involves only two cell types: the primary mesenchymal cells (PMCs) that secrete the calcium carbonate skeleton and the ectodermal cells that provide migratory, positional, and differentiation cues for the PMCs. Perturbations in RA biosynthesis and Hh signaling pathways are thought to be causal for the FAS phenotype in vertebrates. Surprisingly, our results indicate that these pathways are not functionally relevant for the teratogenic effects of ethanol in developing sea urchins. We found that developmental morphology as well as the expression of some ectodermal and PMC genes was delayed by ethanol exposure. Temporal transcriptome analysis revealed significant impacts of ethanol on signaling and metabolic gene expression, and a disruption in the timing of GRN gene expression that includes both delayed and precocious gene expression throughout the specification network. We conclude that the skeletal patterning perturbations in ethanol-treated embryos likely arise from a loss of temporal synchrony within and between the instructive and responsive tissues.
Collapse
Affiliation(s)
| | | | | | - Alexandra T Lion
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA
| | | | - Andrea E Correa
- Universidad de Puerto Rico-Recinto Aguadilla, Puerto Rico, USA
| | | | - Cynthia A Bradham
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA; Biological Design Center, Boston University, Boston, MA, USA.
| |
Collapse
|
4
|
Epigenetics in fetal alcohol spectrum disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:211-239. [PMID: 37019593 DOI: 10.1016/bs.pmbts.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During pregnancy, alcohol abuse and its detrimental effects on developing offspring are major public health, economic and social challenges. The prominent characteristic attributes of alcohol (ethanol) abuse during pregnancy in humans are neurobehavioral impairments in offspring due to damage to the central nervous system (CNS), causing structural and behavioral impairments that are together named fetal alcohol spectrum disorder (FASD). Development-specific alcohol exposure paradigms were established to recapitulate the human FASD phenotypes and establish the underlying mechanisms. These animal studies have offered some critical molecular and cellular underpinnings likely to account for the neurobehavioral impairments associated with prenatal ethanol exposure. Although the pathogenesis of FASD remains unclear, emerging literature proposes that the various genomic and epigenetic components that cause the imbalance in gene expression can significantly contribute to the development of this disease. These studies acknowledged numerous immediate and enduring epigenetic modifications, such as methylation of DNA, post-translational modifications (PTMs) of histone proteins, and regulatory networks related to RNA, using many molecular approaches. Methylated DNA profiles, PTMs of histone proteins, and RNA-regulated expression of genes are essential for synaptic and cognitive behavior. Thus, offering a solution to many neuronal and behavioral impairments reported in FASD. In the current chapter, we review the recent advances in different epigenetic modifications that cause the pathogenesis of FASD. The information discussed can help better explain the pathogenesis of FASD and thereby might provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
|
5
|
Auvinen P, Vehviläinen J, Marjonen H, Modhukur V, Sokka J, Wallén E, Rämö K, Ahola L, Salumets A, Otonkoski T, Skottman H, Ollikainen M, Trokovic R, Kahila H, Kaminen-Ahola N. Chromatin modifier developmental pluripotency associated factor 4 (DPPA4) is a candidate gene for alcohol-induced developmental disorders. BMC Med 2022; 20:495. [PMID: 36581877 PMCID: PMC9801659 DOI: 10.1186/s12916-022-02699-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) affects embryonic development, causing a variable fetal alcohol spectrum disorder (FASD) phenotype with neuronal disorders and birth defects. We hypothesize that early alcohol-induced epigenetic changes disrupt the accurate developmental programming of embryo and consequently cause the complex phenotype of developmental disorders. To explore the etiology of FASD, we collected unique biological samples of 80 severely alcohol-exposed and 100 control newborns at birth. METHODS We performed genome-wide DNA methylation (DNAm) and gene expression analyses of placentas by using microarrays (EPIC, Illumina) and mRNA sequencing, respectively. To test the manifestation of observed PAE-associated DNAm changes in embryonic tissues as well as potential biomarkers for PAE, we examined if the changes can be detected also in white blood cells or buccal epithelial cells of the same newborns by EpiTYPER. To explore the early effects of alcohol on extraembryonic placental tissue, we selected 27 newborns whose mothers had consumed alcohol up to gestational week 7 at maximum to the separate analyses. Furthermore, to explore the effects of early alcohol exposure on embryonic cells, human embryonic stem cells (hESCs) as well as hESCs during differentiation into endodermal, mesodermal, and ectodermal cells were exposed to alcohol in vitro. RESULTS DPPA4, FOXP2, and TACR3 with significantly decreased DNAm were discovered-particularly the regulatory region of DPPA4 in the early alcohol-exposed placentas. When hESCs were exposed to alcohol in vitro, significantly altered regulation of DPPA2, a closely linked heterodimer of DPPA4, was observed. While the regulatory region of DPPA4 was unmethylated in both control and alcohol-exposed hESCs, alcohol-induced decreased DNAm similar to placenta was seen in in vitro differentiated mesodermal and ectodermal cells. Furthermore, common genes with alcohol-associated DNAm changes in placenta and hESCs were linked exclusively to the neurodevelopmental pathways in the enrichment analysis, which emphasizes the value of placental tissue when analyzing the effects of prenatal environment on human development. CONCLUSIONS Our study shows the effects of early alcohol exposure on human embryonic and extraembryonic cells, introduces candidate genes for alcohol-induced developmental disorders, and reveals potential biomarkers for prenatal alcohol exposure.
Collapse
Affiliation(s)
- P Auvinen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - J Vehviläinen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - H Marjonen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - V Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre on Health Technologies, 50411, Tartu, Estonia
| | - J Sokka
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - E Wallén
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - K Rämö
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - L Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - A Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre on Health Technologies, 50411, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, S-171 76, Stockholm, Sweden
| | - T Otonkoski
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - H Skottman
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
| | - M Ollikainen
- Institute for Molecular Medicine, Finland, FIMM, HiLIFE, University of Helsinki, 00290, Helsinki, Finland
| | - R Trokovic
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - H Kahila
- Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - N Kaminen-Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland.
| |
Collapse
|
6
|
Vanhaverbeke M, Attard R, Bartekova M, Ben-Aicha S, Brandenburger T, de Gonzalo-Calvo D, Emanueli C, Farrugia R, Grillari J, Hackl M, Kalocayova B, Martelli F, Scholz M, Wettinger SB, Devaux Y. Peripheral blood RNA biomarkers for cardiovascular disease from bench to bedside: a position paper from the EU-CardioRNA COST action CA17129. Cardiovasc Res 2022; 118:3183-3197. [PMID: 34648023 PMCID: PMC9799060 DOI: 10.1093/cvr/cvab327] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 01/25/2023] Open
Abstract
Despite significant advances in the diagnosis and treatment of cardiovascular diseases, recent calls have emphasized the unmet need to improve precision-based approaches in cardiovascular disease. Although some studies provide preliminary evidence of the diagnostic and prognostic potential of circulating coding and non-coding RNAs, the complex RNA biology and lack of standardization have hampered the translation of these markers into clinical practice. In this position paper of the CardioRNA COST action CA17129, we provide recommendations to standardize the RNA development process in order to catalyse efforts to investigate novel RNAs for clinical use. We list the unmet clinical needs in cardiovascular disease, such as the identification of high-risk patients with ischaemic heart disease or heart failure who require more intensive therapies. The advantages and pitfalls of the different sample types, including RNAs from plasma, extracellular vesicles, and whole blood, are discussed in the sample matrix, together with their respective analytical methods. The effect of patient demographics and highly prevalent comorbidities, such as metabolic disorders, on the expression of the candidate RNA is presented and should be reported in biomarker studies. We discuss the statistical and regulatory aspects to translate a candidate RNA from a research use only assay to an in-vitro diagnostic test for clinical use. Optimal planning of this development track is required, with input from the researcher, statistician, industry, and regulatory partners.
Collapse
Affiliation(s)
- Maarten Vanhaverbeke
- Department of Cardiovascular Medicine, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Ritienne Attard
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida MSD 2080, Malta
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia
- Faculty of Medicine, Institute of Physiology, Comenius University, Sasinkova 2, 81372 Bratislava, Slovakia
| | - Soumaya Ben-Aicha
- Faculty of Medicine, Imperial College London, ICTEM Building, Du Cane Road, London W12 0NN, UK
| | - Timo Brandenburger
- Department of Anesthesiology, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, IRBLleida, University Hospital Arnau de Vilanova and Santa Maria, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Av. de Monforte de Lemos, 28029, Madrid, Spain
| | - Costanza Emanueli
- Faculty of Medicine, Imperial College London, ICTEM Building, Du Cane Road, London W12 0NN, UK
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida MSD 2080, Malta
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU - University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180 Vienna, Austria
| | | | - Barbora Kalocayova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan 20097, Italy
| | - Markus Scholz
- Institute of Medical Informatics, Statistics and Epidemiology, University of Leipzig, Haertelstrasse 16-18, 04107 Leipzig, Germany
| | - Stephanie Bezzina Wettinger
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida MSD 2080, Malta
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Edison, L-1445 Strassen, Luxembourg
| |
Collapse
|
7
|
Lussier AA, Bodnar TS, Weinberg J. Intersection of Epigenetic and Immune Alterations: Implications for Fetal Alcohol Spectrum Disorder and Mental Health. Front Neurosci 2021; 15:788630. [PMID: 34924946 PMCID: PMC8680672 DOI: 10.3389/fnins.2021.788630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Prenatal alcohol exposure can impact virtually all body systems, resulting in a host of structural, neurocognitive, and behavioral abnormalities. Among the adverse impacts associated with prenatal alcohol exposure are alterations in immune function, including an increased incidence of infections and alterations in immune/neuroimmune parameters that last throughout the life-course. Epigenetic patterns are also highly sensitive to prenatal alcohol exposure, with widespread alcohol-related alterations to epigenetic profiles, including changes in DNA methylation, histone modifications, and miRNA expression. Importantly, epigenetic programs are crucial for immune system development, impacting key processes such as immune cell fate, differentiation, and activation. In addition to their role in development, epigenetic mechanisms are emerging as attractive candidates for the biological embedding of environmental factors on immune function and as mediators between early-life exposures and long-term health. Here, following an overview of the impact of prenatal alcohol exposure on immune function and epigenetic patterns, we discuss the potential role for epigenetic mechanisms in reprogramming of immune function and the consequences for health and development. We highlight a range of both clinical and animal studies to provide insights into the array of immune genes impacted by alcohol-related epigenetic reprogramming. Finally, we discuss potential consequences of alcohol-related reprogramming of immune/neuroimmune functions and their effects on the increased susceptibility to mental health disorders. Overall, the collective findings from animal models and clinical studies highlight a compelling relationship between the immune system and epigenetic pathways. These findings have important implications for our understanding of the biological mechanisms underlying the long-term and multisystem effects of prenatal alcohol exposure, laying the groundwork for possible novel interventions and therapeutic strategies to treat individuals prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Fletcher TMD, Mullan B, Novoradovskaya E, Finlay-Jones A. Is 'a little' too much?: An exploration of women's beliefs about alcohol use during pregnancy. Psychol Health 2021:1-19. [PMID: 34658281 DOI: 10.1080/08870446.2021.1991342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Interventions to address alcohol use during pregnancy need to target underlying determinants of the behaviour. Using the theory of planned behaviour as a theoretical framework, the aim of this study was to identify behavioural, normative and control beliefs regarding alcohol use during pregnancy among a sample of women. Design: 435 women completed a 15-minute online questionnaire designed to identify beliefs about alcohol use during pregnancy. Data were categorised according to type of belief and then summarised and described. Results: The majority of respondents saw few advantages of consuming alcohol during pregnancy and believed that most people would disapprove of alcohol use during pregnancy. Although most women endorsed alcohol abstinence during pregnancy, views on the perceived risk of different levels of alcohol use and perceptions of the 'typical' person who drinks while pregnant varied between participants. Conclusion: This work contributes to the understanding of women's beliefs about alcohol use during pregnancy. Future research should explore how women's beliefs inform their decision making about different levels of alcohol use in pregnancy. Additionally, further research or messaging about alcohol use in pregnancy must also consider the potential for contributing to stigmatising beliefs.
Collapse
Affiliation(s)
- Tess M D Fletcher
- FASD Research Australia Centre for Research Excellence, Telethon Kids Institute, Perth, Australia.,Faculty of Health Sciences, Health Psychology & Behavioural Medicine Research Group, Curtin University, Perth, Australia
| | - Barbara Mullan
- Faculty of Health Sciences, Health Psychology & Behavioural Medicine Research Group, Curtin University, Perth, Australia
| | - Elizaveta Novoradovskaya
- Faculty of Health Sciences, Health Psychology & Behavioural Medicine Research Group, Curtin University, Perth, Australia
| | - Amy Finlay-Jones
- FASD Research Australia Centre for Research Excellence, Telethon Kids Institute, Perth, Australia
| |
Collapse
|
9
|
Wallén E, Auvinen P, Kaminen-Ahola N. The Effects of Early Prenatal Alcohol Exposure on Epigenome and Embryonic Development. Genes (Basel) 2021; 12:genes12071095. [PMID: 34356111 PMCID: PMC8303887 DOI: 10.3390/genes12071095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure is one of the most significant causes of developmental disability in the Western world. Maternal alcohol consumption during pregnancy leads to an increased risk of neurological deficits and developmental abnormalities in the fetus. Over the past decade, several human and animal studies have demonstrated that alcohol causes alterations in epigenetic marks, including DNA methylation, histone modifications, and non-coding RNAs. There is an increasing amount of evidence that early pregnancy is a sensitive period for environmental-induced epigenetic changes. It is a dynamic period of epigenetic reprogramming, cell divisions, and DNA replication and, therefore, a particularly interesting period to study the molecular changes caused by alcohol exposure as well as the etiology of alcohol-induced developmental disorders. This article will review the current knowledge about the in vivo and in vitro effects of alcohol exposure on the epigenome, gene regulation, and the phenotype during the first weeks of pregnancy.
Collapse
|
10
|
Rodriguez FD, Coveñas R. Biochemical Mechanisms Associating Alcohol Use Disorders with Cancers. Cancers (Basel) 2021; 13:cancers13143548. [PMID: 34298760 PMCID: PMC8306032 DOI: 10.3390/cancers13143548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Of all yearly deaths attributable to alcohol consumption globally, approximately 12% are due to cancers, representing approximately 0.4 million deceased individuals. Ethanol metabolism disturbs cell biochemistry by targeting the structure and function of essential biomolecules (proteins, nucleic acids, and lipids) and by provoking alterations in cell programming that lead to cancer development and cancer malignancy. A better understanding of the metabolic and cell signaling realm affected by ethanol is paramount to designing effective treatments and preventive actions tailored to specific neoplasias. Abstract The World Health Organization identifies alcohol as a cause of several neoplasias of the oropharynx cavity, esophagus, gastrointestinal tract, larynx, liver, or female breast. We review ethanol’s nonoxidative and oxidative metabolism and one-carbon metabolism that encompasses both redox and transfer reactions that influence crucial cell proliferation machinery. Ethanol favors the uncontrolled production and action of free radicals, which interfere with the maintenance of essential cellular functions. We focus on the generation of protein, DNA, and lipid adducts that interfere with the cellular processes related to growth and differentiation. Ethanol’s effects on stem cells, which are responsible for building and repairing tissues, are reviewed. Cancer stem cells (CSCs) of different origins suffer disturbances related to the expression of cell surface markers, enzymes, and transcription factors after ethanol exposure with the consequent dysregulation of mechanisms related to cancer metastasis or resistance to treatments. Our analysis aims to underline and discuss potential targets that show more sensitivity to ethanol’s action and identify specific metabolic routes and metabolic realms that may be corrected to recover metabolic homeostasis after pharmacological intervention. Specifically, research should pay attention to re-establishing metabolic fluxes by fine-tuning the functioning of specific pathways related to one-carbon metabolism and antioxidant processes.
Collapse
Affiliation(s)
- Francisco D. Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, University of Salamanca, 37007 Salamanca, Spain
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), 37007 Salamanca, Spain;
- Correspondence: ; Tel.: +34-677-510-030
| | - Rafael Coveñas
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), 37007 Salamanca, Spain;
- Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
11
|
Alberry B, Laufer BI, Chater-Diehl E, Singh SM. Epigenetic Impacts of Early Life Stress in Fetal Alcohol Spectrum Disorders Shape the Neurodevelopmental Continuum. Front Mol Neurosci 2021; 14:671891. [PMID: 34149355 PMCID: PMC8209299 DOI: 10.3389/fnmol.2021.671891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
Neurodevelopment in humans is a long, elaborate, and highly coordinated process involving three trimesters of prenatal development followed by decades of postnatal development and maturation. Throughout this period, the brain is highly sensitive and responsive to the external environment, which may provide a range of inputs leading to positive or negative outcomes. Fetal alcohol spectrum disorders (FASD) result from prenatal alcohol exposure (PAE). Although the molecular mechanisms of FASD are not fully characterized, they involve alterations to the regulation of gene expression via epigenetic marks. As in the prenatal stages, the postnatal period of neurodevelopment is also sensitive to environmental inputs. Often this sensitivity is reflected in children facing adverse conditions, such as maternal separation. This exposure to early life stress (ELS) is implicated in the manifestation of various behavioral abnormalities. Most FASD research has focused exclusively on the effect of prenatal ethanol exposure in isolation. Here, we review the research into the effect of prenatal ethanol exposure and ELS, with a focus on the continuum of epigenomic and transcriptomic alterations. Interestingly, a select few experiments have assessed the cumulative effect of prenatal alcohol and postnatal maternal separation stress. Regulatory regions of different sets of genes are affected by both treatments independently, and a unique set of genes are affected by the combination of treatments. Notably, epigenetic and gene expression changes converge at the clustered protocadherin locus and oxidative stress pathway. Functional studies using epigenetic editing may elucidate individual contributions of regulatory regions for hub genes and further profiling efforts may lead to the development of non-invasive methods to identify children at risk. Taken together, the results favor the potential to improve neurodevelopmental outcomes by epigenetic management of children born with FASD using favorable postnatal conditions with or without therapeutic interventions.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Biology, Faculty of Science, The University of Western Ontario, London, ON, Canada
| | - Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States.,Genome Center, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| | - Eric Chater-Diehl
- Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shiva M Singh
- Department of Biology, Faculty of Science, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
12
|
Wiegand A, Kreifelts B, Munk MHJ, Geiselhart N, Ramadori KE, MacIsaac JL, Fallgatter AJ, Kobor MS, Nieratschker V. DNA methylation differences associated with social anxiety disorder and early life adversity. Transl Psychiatry 2021; 11:104. [PMID: 33542190 PMCID: PMC7862482 DOI: 10.1038/s41398-021-01225-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/31/2022] Open
Abstract
Social anxiety disorder (SAD) is a psychiatric disorder characterized by extensive fear in social situations. Multiple genetic and environmental factors are known to contribute to its pathogenesis. One of the main environmental risk factors is early life adversity (ELA). Evidence is emerging that epigenetic mechanisms such as DNA methylation might play an important role in the biological mechanisms underlying SAD and ELA. To investigate the relationship between ELA, DNA methylation, and SAD, we performed an epigenome-wide association study for SAD and ELA examining DNA from whole blood of a cohort of 143 individuals using DNA methylation arrays. We identified two differentially methylated regions (DMRs) associated with SAD located within the genes SLC43A2 and TNXB. As this was the first epigenome-wide association study for SAD, it is worth noting that both genes have previously been associated with panic disorder. Further, we identified two DMRs associated with ELA within the SLC17A3 promoter region and the SIAH3 gene and several DMRs that were associated with the interaction of SAD and ELA. Of these, the regions within C2CD2L and MRPL28 showed the largest difference in DNA methylation. Lastly, we found that two DMRs were associated with both the severity of social anxiety and ELA, however, neither of them was found to mediate the contribution of ELA to SAD later in life. Future studies are needed to replicate our findings in independent cohorts and to investigate the biological pathways underlying these effects.
Collapse
Affiliation(s)
- Ariane Wiegand
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany ,grid.10392.390000 0001 2190 1447Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Benjamin Kreifelts
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Matthias H. J. Munk
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany ,grid.6546.10000 0001 0940 1669Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Nadja Geiselhart
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Katia E. Ramadori
- grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, V5Z 4H4 BC Canada
| | - Julia L. MacIsaac
- grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, V5Z 4H4 BC Canada
| | - Andreas J. Fallgatter
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Michael S. Kobor
- grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, V5Z 4H4 BC Canada
| | - Vanessa Nieratschker
- Department of Psychiatry and Psychotherapy, Eberhard Karls University of Tübingen, Tübingen, Germany. .,Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany.
| |
Collapse
|
13
|
Mohammad S, Page SJ, Sasaki T, Ayvazian N, Rakic P, Kawasawa YI, Hashimoto-Torii K, Torii M. Long-term spatial tracking of cells affected by environmental insults. J Neurodev Disord 2020; 12:38. [PMID: 33327938 PMCID: PMC7745478 DOI: 10.1186/s11689-020-09339-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Harsh environments surrounding fetuses and children can induce cellular damage in the developing brain, increasing the risk of intellectual disability and other neurodevelopmental disorders such as schizophrenia. However, the mechanisms by which early damage leads to disease manifestation in later life remain largely unknown. Previously, we demonstrated that the activation of heat shock (HS) signaling can be utilized as a unique reporter to label the cells that undergo specific molecular/cellular changes upon exposure to environmental insults throughout the body. Since the activation of HS signaling is an acute and transient event, this approach was not intended for long-term tracing of affected cells after the activation has diminished. In the present study, we generated new reporter transgenic mouse lines as a novel tool to achieve systemic and long-term tracking of affected cells and their progeny. Methods The reporter transgenic mouse system was designed so that the activation of HS signaling through HS response element (HSE) drives flippase (FLPo)-flippase recognition target (FRT) recombination-mediated permanent expression of the red fluorescent protein (RFP), tdTomato. With a priority on consistent and efficient assessment of the reporter system, we focused on intraperitoneal (i.p.) injection models of high-dose, short prenatal exposure to alcohol (ethanol) and sodium arsenite (ethanol at 4.0 g/kg/day and sodium arsenite at 5.0 mg/kg/day, at embryonic day (E) 12 and 13). Long-term reporter expression was examined in the brain of reporter mice that were prenatally exposed to these insults. Electrophysiological properties were compared between RFP+ and RFP− cortical neurons in animals prenatally exposed to arsenite. Results We detected RFP+ neurons and glia in the brains of postnatal mice that had been prenatally exposed to alcohol or sodium arsenite. In animals prenatally exposed to sodium arsenite, we also detected reduced excitability in RFP+ cortical neurons. Conclusion The reporter transgenic mice allowed us to trace the cells that once responded to prenatal environmental stress and the progeny derived from these cells long after the exposure in postnatal animals. Tracing of these cells indicates that the impact of prenatal exposure on neural progenitor cells can lead to functional abnormalities in their progeny cells in the postnatal brain. Further studies using more clinically relevant exposure models are warranted to explore this mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-020-09339-w.
Collapse
Affiliation(s)
- Shahid Mohammad
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Stephen J Page
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Toru Sasaki
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Nicholas Ayvazian
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Institute of Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, CT, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
14
|
Tavanasefat H, Li F, Koyano K, Gourtani BK, Marty V, Mulpuri Y, Lee SH, Shin KH, Wong DTW, Xiao X, Spigelman I, Kim Y. Molecular consequences of fetal alcohol exposure on amniotic exosomal miRNAs with functional implications for stem cell potency and differentiation. PLoS One 2020; 15:e0242276. [PMID: 33196678 PMCID: PMC7668603 DOI: 10.1371/journal.pone.0242276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Alcohol (ethanol, EtOH) consumption during pregnancy can result in fetal alcohol spectrum disorders (FASDs), which are characterized by prenatal and postnatal growth restriction and craniofacial dysmorphology. Recently, cell-derived extracellular vesicles, including exosomes and microvesicles containing several species of RNAs (exRNAs), have emerged as a mechanism of cell-to-cell communication. However, EtOH's effects on the biogenesis and function of non-coding exRNAs during fetal development have not been explored. Therefore, we studied the effects of maternal EtOH exposure on the composition of exosomal RNAs in the amniotic fluid (AF) using rat fetal alcohol exposure (FAE) model. Through RNA-Seq analysis we identified and verified AF exosomal miRNAs with differential expression levels specifically associated with maternal EtOH exposure. Uptake of purified FAE AF exosomes by rBMSCs resulted in significant alteration of molecular markers associated with osteogenic differentiation of rBMSCs. We also determined putative functional roles for AF exosomal miRNAs (miR-199a-3p, miR-214-3p and let-7g) that are dysregulated by FAE in osteogenic differentiation of rBMSCs. Our results demonstrate that FAE alters AF exosomal miRNAs and that exosomal transfer of dysregulated miRNAs has significant molecular effects on stem cell regulation and differentiation. Our results further suggest the usefulness of assessing molecular alterations in AF exRNAs to study the mechanisms of FAE teratogenesis that should be further investigated by using an in vivo model.
Collapse
Affiliation(s)
- Honey Tavanasefat
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, UCLA, Los Angeles, California, United States of America
- CSUN-UCLA Stem Cell Research Bridge Program, Department of Biology, California State University at Northridge, Northridge, California, United States of America
| | - Feng Li
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Kikuye Koyano
- Department of Integrative Biology and Physiology, Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Bahar Khalilian Gourtani
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, UCLA, Los Angeles, California, United States of America
| | - Vincent Marty
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Yatendra Mulpuri
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - David T. W. Wong
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Igor Spigelman
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
| | - Yong Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, UCLA, Los Angeles, California, United States of America
- Division of Oral Biology & Medicine, UCLA School of Dentistry, Los Angeles, California, United States of America
- UCLA Broad Stem Cell Research Center, Los Angeles, California, United States of America
| |
Collapse
|
15
|
Kaminen-Ahola N. Fetal alcohol spectrum disorders: Genetic and epigenetic mechanisms. Prenat Diagn 2020; 40:1185-1192. [PMID: 32386259 DOI: 10.1002/pd.5731] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/26/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are a consequence of prenatal alcohol exposure (PAE). The etiology of the complex FASD phenotype with growth deficit, birth defects, and neurodevelopmental impairments is under extensive research. Both genetic and environmental factors contribute to the wide phenotype: chromosomal rearrangements, risk and protective alleles, environmental-induced epigenetic alterations as well as gene-environment interactions are all involved. Understanding the molecular mechanisms of PAE can provide tools for prevention or intervention of the alcohol-induced developmental disorders in the future. By revealing the alcohol-induced genetic and epigenetic alterations which associate with the variable FASD phenotypes, it is possible to identify biomarkers for the disorder. This would enable early diagnoses and personalized support for development of the affected child.
Collapse
Affiliation(s)
- Nina Kaminen-Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Alberry BLJ, Castellani CA, Singh SM. Hippocampal transcriptome analysis following maternal separation implicates altered RNA processing in a mouse model of fetal alcohol spectrum disorder. J Neurodev Disord 2020; 12:15. [PMID: 32416732 PMCID: PMC7231420 DOI: 10.1186/s11689-020-09316-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/16/2020] [Indexed: 01/05/2023] Open
Abstract
Background Fetal alcohol spectrum disorders (FASD) are common, seen in 1–5% of the population in the USA and Canada. Children diagnosed with FASD are not likely to remain with their biological parents, facing early maternal separation and foster placements throughout childhood. Methods We model FASD in mice via prenatal alcohol exposure and further induce early life stress through maternal separation. We use RNA-seq followed by clustering of expression profiles through weighted gene co-expression network analysis (WGCNA) to analyze transcriptomic changes that result from the treatments. We use reverse transcription qPCR to validate these changes in the mouse hippocampus. Results We report an association between adult hippocampal gene expression and prenatal ethanol exposure followed by postnatal separation stress that is related to behavioral changes. Expression profile clustering using WGCNA identifies a set of transcripts, module 19, associated with anxiety-like behavior (r = 0.79, p = 0.002) as well as treatment group (r = 0.68, p = 0.015). Genes in this module are overrepresented by genes involved in transcriptional regulation and other pathways related to neurodevelopment. Interestingly, one member of this module, Polr2a, polymerase (RNA) II (DNA directed) polypeptide A, is downregulated by the combination of prenatal ethanol and postnatal stress in an RNA-Seq experiment and qPCR validation (q = 2e−12, p = 0.004, respectively). Conclusions Together, transcriptional control in the hippocampus is implicated as a potential underlying mechanism leading to anxiety-like behavior via environmental insults. Further research is required to elucidate the mechanism involved and use this insight towards early diagnosis and amelioration strategies involving children born with FASD.
Collapse
Affiliation(s)
- Bonnie L J Alberry
- Department of Biology, Western University, 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - Christina A Castellani
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Shiva M Singh
- Department of Biology, Western University, 1151 Richmond St, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
17
|
de Angelis C, Nardone A, Garifalos F, Pivonello C, Sansone A, Conforti A, Di Dato C, Sirico F, Alviggi C, Isidori A, Colao A, Pivonello R. Smoke, alcohol and drug addiction and female fertility. Reprod Biol Endocrinol 2020; 18:21. [PMID: 32164734 PMCID: PMC7069005 DOI: 10.1186/s12958-020-0567-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Considerable interest has been gathered on the relevant impact of preventable factors, including incorrect lifestyle and unhealthy habits, on female fertility. Smoking, alcohol and addictive drugs consumption represent a major concern, given the broad range of diseases which might be favored or exacerbated by these dependable attitudes. Despite the well-characterized effects of prenatal exposure on pregnancy outcomes and fetus health, a substantial proportion of women of reproductive age is still concerned with these habits. At present, the impact of smoke, alcohol and addictive drugs on women fertility, and, particularly, the specific targets and underlying mechanisms, are still poorly understood or debated, mainly due to the scarcity of well-designed studies, and to numerous biases. OBJECTIVE The current review will provide a comprehensive overview of clinical and experimental studies in humans and animals addressing the impact of smoke, alcohol and addictive drugs on female fertility, by also embracing effects on ovary, oviduct, and uterus, with particular reference to primary endpoints such as ovarian reserve, steroidogenesis, ovulation and menstrual cycle, oviduct function and uterus receptivity and implantation. A brief focus on polycystic ovary syndrome and endometriosis will be also included. METHODS A Pubmed literature search was performed with selected keywords; articles were individually retrieved by each author. No limitation was set for publication date. Articles in languages other than English were excluded. Additional articles were retrieved from references list of selected manuscripts. RESULTS AND CONCLUSIONS Currently, the most consistent evidences of a detrimental effect of smoke, alcohol and addictive drugs on specific domains of the female reproductive function are provided by experimental studies in animals. Overall, clinical studies suggest that smoking is associated to decreased fertility, although causal inference should be further demonstrated. Studies addressing the effect of alcohol consumption on female fertility provide conflicting results, although the majority reported lack of a correlation. Extremely scarce studies investigated the effects of addictive drugs on female fertility, and the specific actions of selected drugs have been difficult to address, due to multidrug consumption.
Collapse
Affiliation(s)
- Cristina de Angelis
- I.O.S. & COLEMAN Srl, Naples, Italy
- grid.4691.a0000 0001 0790 385XDipartimento di Medicina Clinica e Chirurgia, Università “Federico II” di Napoli, Via Sergio Pansini 5, 80131 Naples, Italy
- grid.4691.a0000 0001 0790 385XFERTISEXCARES Centro di Andrologia, Medicina della Riproduzione e della Sessualità Maschile e Femminile, Università “Federico II” di Napoli, Naples, Italy
- grid.4691.a0000 0001 0790 385XDipartimento di Sanità Pubblica, Università “Federico II” di Napoli, Naples, Italy
| | - Antonio Nardone
- grid.4691.a0000 0001 0790 385XDipartimento di Sanità Pubblica, Università “Federico II” di Napoli, Naples, Italy
| | - Francesco Garifalos
- grid.4691.a0000 0001 0790 385XDipartimento di Medicina Clinica e Chirurgia, Università “Federico II” di Napoli, Via Sergio Pansini 5, 80131 Naples, Italy
- grid.4691.a0000 0001 0790 385XFERTISEXCARES Centro di Andrologia, Medicina della Riproduzione e della Sessualità Maschile e Femminile, Università “Federico II” di Napoli, Naples, Italy
| | - Claudia Pivonello
- grid.4691.a0000 0001 0790 385XDipartimento di Medicina Clinica e Chirurgia, Università “Federico II” di Napoli, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Andrea Sansone
- grid.7841.aDepartment of Experimental Medicine, Faculty of Medicine and Dentistry, University of Rome “Sapienza”, viale Regina Elena 324, 00162 Roma, Italy
| | - Alessandro Conforti
- grid.4691.a0000 0001 0790 385XDepartment of Neuroscience, Reproductive Medicine, Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Carla Di Dato
- grid.7841.aDepartment of Experimental Medicine, Faculty of Medicine and Dentistry, University of Rome “Sapienza”, viale Regina Elena 324, 00162 Roma, Italy
| | - Felice Sirico
- grid.4691.a0000 0001 0790 385XDipartimento di Sanità Pubblica, Università “Federico II” di Napoli, Naples, Italy
| | - Carlo Alviggi
- grid.4691.a0000 0001 0790 385XDepartment of Neuroscience, Reproductive Medicine, Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Andrea Isidori
- grid.7841.aDepartment of Experimental Medicine, Faculty of Medicine and Dentistry, University of Rome “Sapienza”, viale Regina Elena 324, 00162 Roma, Italy
| | - Annamaria Colao
- grid.4691.a0000 0001 0790 385XDipartimento di Medicina Clinica e Chirurgia, Università “Federico II” di Napoli, Via Sergio Pansini 5, 80131 Naples, Italy
- grid.4691.a0000 0001 0790 385XFERTISEXCARES Centro di Andrologia, Medicina della Riproduzione e della Sessualità Maschile e Femminile, Università “Federico II” di Napoli, Naples, Italy
- grid.4691.a0000 0001 0790 385XCattedra Unesco “Educazione alla salute e allo sviluppo sostenibile”, Università “Federico II” di Napoli, Naples, Italy
| | - Rosario Pivonello
- grid.4691.a0000 0001 0790 385XDipartimento di Medicina Clinica e Chirurgia, Università “Federico II” di Napoli, Via Sergio Pansini 5, 80131 Naples, Italy
- grid.4691.a0000 0001 0790 385XFERTISEXCARES Centro di Andrologia, Medicina della Riproduzione e della Sessualità Maschile e Femminile, Università “Federico II” di Napoli, Naples, Italy
- grid.4691.a0000 0001 0790 385XCattedra Unesco “Educazione alla salute e allo sviluppo sostenibile”, Università “Federico II” di Napoli, Naples, Italy
| |
Collapse
|
18
|
Serio RN, Gudas LJ. Modification of stem cell states by alcohol and acetaldehyde. Chem Biol Interact 2019; 316:108919. [PMID: 31846616 PMCID: PMC7036011 DOI: 10.1016/j.cbi.2019.108919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/13/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022]
Abstract
Ethanol (EtOH) is a recreationally ingested compound that is both teratogenic and carcinogenic in humans. Because of its abundant consumption worldwide and the vital role of stem cells in the formation of birth defects and cancers, delineating the effects of EtOH on stem cell function is currently an active and urgent pursuit of scientific investigation to explicate some of the mechanisms contributing to EtOH toxicity. Stem cells represent a primordial, undifferentiated phase of development; thus encroachment on normal physiologic processes of differentiation into terminal lineages by EtOH can greatly alter the function of progenitors and terminally differentiated cells, leading to pathological consequences that manifest as fetal alcohol spectrum disorders and cancers. In this review we explore the disruptive role of EtOH in differentiation of stem cells. Our primary objective is to elucidate the mechanisms by which EtOH alters differentiation-related gene expression and lineage specifications, thus modifying stem cells to promote pathological outcomes. We additionally review the effects of a reactive metabolite of EtOH, acetaldehyde (AcH), in causing both differentiation defects in stem cells as well as genomic damage that incites cellular aging and carcinogenesis.
Collapse
Affiliation(s)
- Ryan N Serio
- Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, USA.
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, USA; Department of Pharmacology, Weill Cornell Medical College of Cornell University, USA.
| |
Collapse
|
19
|
Bendre M, Granholm L, Drennan R, Meyer A, Yan L, Nilsson KW, Nylander I, Comasco E. Early life stress and voluntary alcohol consumption in relation to Maoa methylation in male rats. Alcohol 2019; 79:7-16. [PMID: 30414913 DOI: 10.1016/j.alcohol.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 01/22/2023]
Abstract
Early life stress (ELS) or alcohol consumption can influence DNA methylation and affect gene expression. Monoamine oxidase A (Maoa) encodes the enzyme that metabolizes monoaminergic neurotransmitters crucial for the stress response, alcohol reward, and reinforcement. Previously, we reported lower Maoa expression in the nucleus accumbens and dorsal striatum of male rats exposed to ELS during the first three postnatal weeks, and to voluntary alcohol consumption in adulthood, compared with controls. The present study continued to investigate the effect of ELS and alcohol consumption on Maoa methylation, and its relation to Maoa expression in these animals. We selected candidate CpGs after performing next-generation bisulfite sequencing of the Maoa promoter, intron 1-5, and exons 5 and 6, together composed of 107 CpGs (5'-cytosine-phosphate-guanosine-3'), in a subgroup of rats. Pyrosequencing was used to analyze the methylation of 10 candidate CpGs in the promoter and intron 1 in the entire sample. ELS and alcohol displayed an interactive effect on CpG-specific methylation in the dorsal striatum. CpG-specific methylation correlated with Maoa expression, corticosterone levels, and alcohol consumption in a brain region-specific manner. CpG-specific methylation in the Maoa promoter was a potential moderator of the interaction of ELS with alcohol consumption on Maoa expression in the NAc. However, the findings were sparse, did not survive correction for multiple testing, and the magnitude of differences in methylation levels was small. In conclusion, CpG-specific Maoa methylation in the promoter and intron 1 may associate with ELS, alcohol consumption, and Maoa expression in reward-related brain regions.
Collapse
|
20
|
Abstract
Alcohol exposure during pregnancy results in impaired growth, stillbirth, and fetal alcohol spectrum disorder. Fetal alcohol deficits are lifelong issues with no current treatment or established diagnostic or therapeutic tools to prevent and/or ameliorate some of these adverse outcomes. Despite the recommendation to abstain, almost half of the women consume alcohol in pregnancy in the United States. This review focuses on the trends in prenatal alcohol exposure, implications for maternal and fetal health, and evidence suggesting that preconception and the prenatal period provide a window of opportunity to intervene, mitigate, and ideally curtail the lifetime effects of fetal alcohol spectrum disorder.
Collapse
|
21
|
Lunde‐Young R, Ramirez J, Naik V, Orzabal M, Lee J, Konganti K, Hillhouse A, Threadgill D, Ramadoss J. Hippocampal transcriptome reveals novel targets of FASD pathogenesis. Brain Behav 2019; 9:e01334. [PMID: 31140755 PMCID: PMC6625466 DOI: 10.1002/brb3.1334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/01/2019] [Accepted: 05/06/2019] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Prenatal alcohol exposure can contribute to fetal alcohol spectrum disorders (FASD), characterized by a myriad of developmental impairments affecting behavior and cognition. Studies show that many of these functional impairments are associated with the hippocampus, a structure exhibiting exquisite vulnerability to developmental alcohol exposure and critically implicated in learning and memory; however, mechanisms underlying alcohol-induced hippocampal deficits remain poorly understood. By utilizing a high-throughput RNA-sequencing (RNA-seq) approach to address the neurobiological and molecular basis of prenatal alcohol-induced hippocampal functional deficits, we hypothesized that chronic binge prenatal alcohol exposure alters gene expression and global molecular pathways in the fetal hippocampus. METHODS Timed-pregnant Sprague-Dawley rats were randomly assigned to a pair-fed control (PF) or binge alcohol (ALC) treatment group on gestational day (GD) 4. ALC dams acclimatized from GDs 5-10 with a daily treatment of 4.5 g/kg alcohol and subsequently received 6 g/kg on GDs 11-20. PF dams received a once daily maltose dextrin gavage on GDs 5-20, isocalorically matching ALC counterparts. On GD 21, bilateral hippocampi were dissected, flash frozen, and stored at -80° C. Total RNA was then isolated from homogenized tissues. Samples were normalized to ~4nM and pooled equally. Sequencing was performed by Illumina NextSeq 500 on a 75 cycle, single-end sequencing run. RESULTS RNA-seq identified 13,388 genes, of these, 76 genes showed a significant difference (p < 0.05, log2 fold change ≥2) in expression between the PF and ALC groups. Forty-nine genes showed sex-dependent dysregulation; IPA analysis showed among female offspring, dysregulated pathways included proline and citrulline biosynthesis, whereas in males, xenobiotic metabolism signaling and alaninine biosynthesis etc. were altered. CONCLUSION We conclude that chronic binge alcohol exposure during pregnancy dysregulates fetal hippocampal gene expression in a sex-specific manner. Identification of subtle, transcriptome-level dysregulation in hippocampal molecular pathways offers potential mechanistic insights underlying FASD pathogenesis.
Collapse
Affiliation(s)
- Raine Lunde‐Young
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Josue Ramirez
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Vishal Naik
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Marcus Orzabal
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Jehoon Lee
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Kranti Konganti
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Andrew Hillhouse
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - David Threadgill
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| | - Jayanth Ramadoss
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexas
| |
Collapse
|
22
|
Di Rocco G, Baldari S, Pani G, Toietta G. Stem cells under the influence of alcohol: effects of ethanol consumption on stem/progenitor cells. Cell Mol Life Sci 2019; 76:231-244. [PMID: 30306211 PMCID: PMC6339663 DOI: 10.1007/s00018-018-2931-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
Stem cells drive embryonic and fetal development. In several adult tissues, they retain the ability to self-renew and differentiate into a variety of specialized cells, thus contributing to tissue homeostasis and repair throughout life span. Alcohol consumption is associated with an increased risk for several diseases and conditions. Growing and developing tissues are particularly vulnerable to alcohol's influence, suggesting that stem- and progenitor-cell function could be affected. Accordingly, recent studies have revealed the possible relevance of alcohol exposure in impairing stem-cell properties, consequently affecting organ development and injury response in different tissues. Here, we review the main studies describing the effects of alcohol on different types of progenitor/stem cells including neuronal, hepatic, intestinal and adventitial progenitor cells, bone-marrow-derived stromal cell, dental pulp, embryonic and hematopoietic stem cells, and tumor-initiating cells. A better understanding of the nature of the cellular damage induced by chronic and episodic heavy (binge) drinking is critical for the improvement of current therapeutic strategies designed to treat patients suffering from alcohol-related disorders.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Giovambattista Pani
- Institute of General Pathology, Laboratory of Cell Signaling, Catholic University Medical School, Largo F. Vito 1, 00168, Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
23
|
Down-regulation of HIF-1α inhibits the proliferation, migration, and invasion of gastric cancer by inhibiting PI3K/AKT pathway and VEGF expression. Biosci Rep 2018; 38:BSR20180741. [PMID: 29899167 PMCID: PMC6435555 DOI: 10.1042/bsr20180741] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
In view of the high incidence of gastric cancer and the functions of hypoxia-inducible factor 1α (HIF-1α), our study aimed to investigate the functionality of HIF-1α in gastric cancer, and to explore the diagnostic and prognostic values of HIF-1α for this disease. Expression of HIF-1α in tumor tissues and adjacent healthy tissues as well as serum collected from both gastric cancer patients and normal healthy controls was detected by qRT-PCR. Survival analysis was performed using Kaplan–Meier method. HIF-1α siRNA silencing cell lines were established. Effects of HIF-1α siRNA silencing as well as PI3K activator sc3036 on proliferation, migration, and invasion of gastric cancer cells were detected by Cell counting kit (CCK-8) assay, and Transwell migration and invasion assay. Effects of HIF-1α siRNA silencing on AKT and VEGF were detected by Western blot. Expression of HIF-1α was significantly down-regulated in tumor tissues than in adjacent healthy tissues in most gastric cancer patients. Serum levels of HIF-1α were also higher in gastric cancer patients than in normal healthy people. Serum HIF-1α showed promising diagnostic and prognostic values for gastric cancer. HIF-1α siRNA silencing inhibited the proliferation, migration, and invasion of gastric cancer cells, while PI3K activator sc3036 treatment reduced those inhibitory effects. Down-regulation of HIF-1α can inhibit the proliferation, migration, and invasion of gastric cancer possibly by inhibiting PI3K/AKT pathway and VEGF expression.
Collapse
|
24
|
Apáti Á, Varga N, Berecz T, Erdei Z, Homolya L, Sarkadi B. Application of human pluripotent stem cells and pluripotent stem cell-derived cellular models for assessing drug toxicity. Expert Opin Drug Metab Toxicol 2018; 15:61-75. [PMID: 30526128 DOI: 10.1080/17425255.2019.1558207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Human pluripotent stem cells (hPSCs) are capable of differentiating into all types of cells in the body and so provide suitable toxicology screening systems even for hard-to-obtain human tissues. Since hPSCs can also be generated from differentiated cells and current gene editing technologies allow targeted genome modifications, hPSCs can be applied for drug toxicity screening both in normal and disease-specific models. Targeted hPSC differentiation is still a challenge but cardiac, neuronal or liver cells, and complex cellular models are already available for practical applications. Areas covered: The authors review new gene-editing and cell-biology technologies to generate sensitive toxicity screening systems based on hPSCs. Then the authors present the use of undifferentiated hPSCs for examining embryonic toxicity and discuss drug screening possibilities in hPSC-derived models. The authors focus on the application of human cardiomyocytes, hepatocytes, and neural cultures in toxicity testing, and discuss the recent possibilities for drug screening in a 'body-on-a-chip' model system. Expert opinion: hPSCs and their genetically engineered derivatives provide new possibilities to investigate drug toxicity in human tissues. The key issues in this regard are still the selection and generation of proper model systems, and the interpretation of the results in understanding in vivo drug effects.
Collapse
Affiliation(s)
- Ágota Apáti
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Nóra Varga
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Tünde Berecz
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Zsuzsa Erdei
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - László Homolya
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Balázs Sarkadi
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| |
Collapse
|
25
|
Luz AL, Tokar EJ. Pluripotent Stem Cells in Developmental Toxicity Testing: A Review of Methodological Advances. Toxicol Sci 2018; 165:31-39. [PMID: 30169765 PMCID: PMC6111785 DOI: 10.1093/toxsci/kfy174] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Millions of children are born each year with a birth defect. Many of these defects are caused by environmental factors, although the underlying etiology is often unknown. In vivo mammalian models are frequently used to determine if a chemical poses a risk to the developing fetus. However, there are over 80 000 chemicals registered for use in the United States, many of which have undergone little safety testing, necessitating the need for higher-throughput methods to assess developmental toxicity. Pluripotent stem cells (PSCs) are an ideal in vitro model to investigate developmental toxicity as they possess the capacity to differentiate into nearly any cell type in the human body. Indeed, a burst of research has occurred in the field of stem cell toxicology over the past decade, which has resulted in numerous methodological advances that utilize both mouse and human PSCs, as well as cutting-edge technology in the fields of metabolomics, transcriptomics, transgenics, and high-throughput imaging. Here, we review the wide array of approaches used to detect developmental toxicants, suggest areas for further research, and highlight critical aspects of stem cell biology that should be considered when utilizing PSCs in developmental toxicity testing.
Collapse
Affiliation(s)
- Anthony L Luz
- Stem Cell Toxicology Group, National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Erik J Tokar
- Stem Cell Toxicology Group, National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| |
Collapse
|
26
|
Reece AS, Wang W, Hulse GK. Pathways from epigenomics and glycobiology towards novel biomarkers of addiction and its radical cure. Med Hypotheses 2018; 116:10-21. [PMID: 29857889 DOI: 10.1016/j.mehy.2018.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 03/25/2018] [Accepted: 04/11/2018] [Indexed: 12/12/2022]
Abstract
The recent demonstration that addiction-relevant neuronal ensembles defined by known master transcription factors and their connectome is networked throughout mesocorticolimbic reward circuits and resonates harmonically at known frequencies implies that single-cell pan-omics techniques can improve our understanding of Substance Use Disorders (SUD's). Application of machine learning algorithms to such data could find diagnostic utility as biomarkers both to define the presence of the disorder and to quantitate its severity and find myriad applications in a developmental pipeline towards therapeutics and cure. Recent epigenomic studies have uncovered a wealth of clinically important data relating to synapse-nucleus signalling, memory storage, lineage-fate determination and cellular control and are contributing greatly to our understanding of all SUD's. Epigenetics interacts extensively with glycobiology. Glycans decorate DNA, RNA and many circulating critical proteins particularly immunoglobulins. Glycosylation is emerging as a major information-laden post-translational protein modification with documented application for biomarker development. The integration of these two emerging cutting-edge technologies provides a powerful and fertile algorithmic-bioinformatic space for the development both of SUD biomarkers and novel cutting edge therapeutics. HYPOTHESES These lines of evidence provide fertile ground for hypotheses relating to both diagnosis and treatment. They suggest that biomarkers derived from epigenomics complemented by glycobiology may potentially provide a bedside diagnostic tool which could be developed into a clinically useful biomarker to gauge both the presence and the severity of SUD's. Moreover they suggest that modern information-based therapeutics acting on the epigenome, via RNA interference or by DNA antisense oligonucleotides may provide a novel 21st century therapeutic development pipeline towards the radical cure of addictive disorders. Such techniques could be focussed and potentiated by neurotrophic vectors or the application of interfering electric or magnetic fields deep in the medial temporal lobes of the brain.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, Western Australia 6009, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
| | - Wei Wang
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, Western Australia 6009, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| |
Collapse
|
27
|
Li J, Wang Z, Qiu W, Yang JJ, Wang Q, Chen S, Pan H. The effect of interaction between EtOH dosage and exposure time on gene expression in DPSC. Genomics 2018; 111:500-507. [PMID: 29596963 DOI: 10.1016/j.ygeno.2018.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/11/2018] [Accepted: 03/07/2018] [Indexed: 12/17/2022]
Abstract
Alcohol (EtOH) dosage and exposure time can affect gene expression. However, whether there exists synergistic effect is unknown. Here, we analyzed the hDPSC gene microarray dataset GSE57255 downloaded from Gene Expression Omnibus and found that the interaction between EtOH dosage and exposure time on gene expression are statistically significant for two probes: 201917_s_at near gene SLC25A36 and 217649_at near gene ZFAND5. GeneMania showed that SLC25A36 and ZFAND5 were related to 20 genes, three of which had alcohol-related functions. WebGestalt revealed that the 22 genes were enriched in 10 KEGG pathways, four of which are related to alcoholic diseases. We explored the possible nonlinear interaction effect and got 172 gene probes with significant p-values. However, no significantly enriched pathways based on the 172 probes were detected. Our analyses indicated a possible molecular mechanism that could help explain why alcohol consumption has both deleterious and beneficial effects on human health.
Collapse
Affiliation(s)
- Jianqiang Li
- School of Software Engineering, Beijing University of Technology, Beijing, China
| | - Zhirui Wang
- School of Software Engineering, Beijing University of Technology, Beijing, China
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Ji-Jiang Yang
- Tsinghua National Laboratory for Information Science and Technology, Tsinghua University, Beijing, China.
| | - Qing Wang
- Tsinghua National Laboratory for Information Science and Technology, Tsinghua University, Beijing, China
| | - Shi Chen
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing, China
| | - Hui Pan
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
28
|
Xiong Z, Ren S, Chen H, Liu Y, Huang C, Zhang YL, Odera JO, Chen T, Kist R, Peters H, Garman K, Sun Z, Chen X. PAX9 regulates squamous cell differentiation and carcinogenesis in the oro-oesophageal epithelium. J Pathol 2018; 244:164-175. [PMID: 29055049 PMCID: PMC5842438 DOI: 10.1002/path.4998] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/05/2017] [Accepted: 10/10/2017] [Indexed: 12/28/2022]
Abstract
PAX9 is a transcription factor of the PAX family characterized by a DNA-binding paired domain. Previous studies have suggested a potential role of PAX9 in squamous cell differentiation and carcinogenesis of the oro-oesophageal epithelium. However, its functional roles in differentiation and carcinogenesis remain unclear. In this study, Pax9 deficiency in mouse oesophagus promoted cell proliferation, delayed cell differentiation, and altered the global gene expression profile. Ethanol exposure downregulated PAX9 expression in human oesophageal epithelial cells in vitro and mouse forestomach and tongue in vivo. We further showed that PAX9 was downregulated in human oro-oesophageal squamous cell carcinoma (OESCC), and its downregulation was associated with alcohol drinking and promoter hypermethylation. Moreover, ad libitum feeding with a liquid diet containing ethanol for 40 weeks or Pax9 deficiency promoted N-nitrosomethylbenzylamine-induced squamous cell carcinogenesis in mouse tongue, oesophagus, and forestomach. In conclusion, PAX9 regulates squamous cell differentiation in the oro-oesophageal epithelium. Alcohol drinking and promoter hypermethylation are associated with PAX9 silencing in human OESCC. PAX9 downregulation may contribute to alcohol-associated oro-oesophageal squamous cell carcinogenesis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zhaohui Xiong
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Shuang Ren
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Hao Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Yao Liu
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Caizhi Huang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Yawan Lyvia Zhang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Joab Otieno Odera
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Tong Chen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, 410 West 12 Avenue, Columbus, OH 43210, USA
| | - Ralf Kist
- Centre for Oral Health Research, School of Dental Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4BW, UK
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Heiko Peters
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Katherine Garman
- Division of Gastroenterology, Department of Medicine, Duke University, DUMC 3913, Durham, NC 27710, USA
| | - Zheng Sun
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
| | - Xiaoxin Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
29
|
Manto M, Perrotta G. Toxic-induced cerebellar syndrome: from the fetal period to the elderly. HANDBOOK OF CLINICAL NEUROLOGY 2018; 155:333-352. [DOI: 10.1016/b978-0-444-64189-2.00022-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Miozzo F, Arnould H, de Thonel A, Schang AL, Sabéran-Djoneidi D, Baudry A, Schneider B, Mezger V. Alcohol exposure promotes DNA methyltransferase DNMT3A upregulation through reactive oxygen species-dependent mechanisms. Cell Stress Chaperones 2018; 23:115-126. [PMID: 28712054 PMCID: PMC5741586 DOI: 10.1007/s12192-017-0829-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022] Open
Abstract
Abundant evidence has accumulated showing that fetal alcohol exposure broadly modifies DNA methylation profiles in the brain. DNA methyltransferases (DNMTs), the enzymes responsible for DNA methylation, are likely implicated in this process. However, their regulation by ethanol exposure has been poorly addressed. Here, we show that alcohol exposure modulates DNMT protein levels through multiple mechanisms. Using a neural precursor cell line and primary mouse embryonic fibroblasts (MEFs), we found that ethanol exposure augments the levels of Dnmt3a, Dnmt3b, and Dnmt3l transcripts. We also unveil similar elevation of mRNA levels for other epigenetic actors upon ethanol exposure, among which the induction of lysine demethylase Kdm6a shows heat shock factor dependency. Furthermore, we show that ethanol exposure leads to specific increase in DNMT3A protein levels. This elevation not only relies on the upregulation of Dnmt3a mRNA but also depends on posttranscriptional mechanisms that are mediated by NADPH oxidase-dependent production of reactive oxygen species (ROS). Altogether, our work underlines complex regulation of epigenetic actors in response to alcohol exposure at both transcriptional and posttranscriptional levels. Notably, the upregulation of DNMT3A emerges as a prominent molecular event triggered by ethanol, driven by the generation of ROS.
Collapse
Affiliation(s)
- Federico Miozzo
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
- Department of Genetics and Evolution, Sciences III, University of Geneva, Geneva, Switzerland
| | - Hélène Arnould
- INSERM UMR-S1124, Paris Cedex 6, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France
| | - Aurélie de Thonel
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
| | - Anne-Laure Schang
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
- UMR CNRS 8638-Chimie Toxicologie Analytique et Cellulaire, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
| | - Anne Baudry
- INSERM UMR-S1124, Paris Cedex 6, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France
| | - Benoît Schneider
- INSERM UMR-S1124, Paris Cedex 6, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France.
| | - Valérie Mezger
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France.
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France.
- Département Hospitalo-Universitaire PROTECT, Paris, France.
| |
Collapse
|
31
|
Rea M, Gripshover T, Fondufe-Mittendorf Y. Selective inhibition of CTCF binding by iAs directs TET-mediated reprogramming of 5-hydroxymethylation patterns in iAs-transformed cells. Toxicol Appl Pharmacol 2018; 338:124-133. [PMID: 29175454 PMCID: PMC5738917 DOI: 10.1016/j.taap.2017.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/17/2017] [Accepted: 11/19/2017] [Indexed: 12/22/2022]
Abstract
Methylation at cytosine (5mC) is a fundamental epigenetic DNA modification recently associated with iAs-mediated carcinogenesis. In contrast, the role of 5-hydroxymethylcytosine (5hmC), the oxidation product of 5mC in iAs-mediated carcinogenesis is unknown. Here we assess the hydroxymethylome in iAs-transformed cells, showing that dynamic modulation of hydroxymethylated DNA is associated with specific transcriptional networks. Moreover, this pathologic iAs-mediated carcinogenesis is characterized by a shift toward a higher hydroxymethylation pattern genome-wide. At specific promoters, hydroxymethylation correlated with increased gene expression. Furthermore, this increase in hydroxymethylation occurs concurrently with an upregulation of ten-eleven translocation (TET) enzymes that oxidize 5-methylcytosine (5mC) in DNA. To gain an understanding into how iAs might impact TET expression, we found that iAs inhibits the binding of CTCF at the proximal, weak CTCF binding sites of the TET1 and TET2 gene promoters and enhances CTCF binding at the stronger distal binding site. Further analyses suggest that this distal site acts as an enhancer, thus high CTCF occupancy at the enhancer region of TET1 and TET2 possibly drives their high expression in iAs-transformed cells. These results have major implications in understanding the impact of differential CTCF binding, genome architecture and its consequences in iAs-mediated pathogenesis.
Collapse
Affiliation(s)
- Matthew Rea
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Tyler Gripshover
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA; Eastern Kentucky University, Richmond, KY 40475, USA
| | - Yvonne Fondufe-Mittendorf
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
32
|
Kirischuk S, Sinning A, Blanquie O, Yang JW, Luhmann HJ, Kilb W. Modulation of Neocortical Development by Early Neuronal Activity: Physiology and Pathophysiology. Front Cell Neurosci 2017; 11:379. [PMID: 29238291 PMCID: PMC5712676 DOI: 10.3389/fncel.2017.00379] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022] Open
Abstract
Animal and human studies revealed that patterned neuronal activity is an inherent feature of developing nervous systems. This review summarizes our current knowledge about the mechanisms generating early electrical activity patterns and their impact on structural and functional development of the cerebral cortex. All neocortical areas display distinct spontaneous and sensory-driven neuronal activity patterns already at early phases of development. At embryonic stages, intermittent spontaneous activity is synchronized within small neuronal networks, becoming more complex with further development. This transition is accompanied by a gradual shift from electrical to chemical synaptic transmission, with a particular role of non-synaptic tonic currents before the onset of phasic synaptic activity. In this review article we first describe functional impacts of classical neurotransmitters (GABA, glutamate) and modulatory systems (e.g., acetylcholine, ACh) on early neuronal activities in the neocortex with special emphasis on electrical synapses, nonsynaptic and synaptic currents. Early neuronal activity influences probably all developmental processes and is crucial for the proper formation of neuronal circuits. In the second part of our review, we illustrate how specific activity patterns might interfere with distinct neurodevelopmental processes like proliferation, migration, axonal and dendritic sprouting, synapse formation and neurotransmitter specification. Finally, we present evidence that transient alterations in neuronal activity during restricted perinatal periods can lead to persistent changes in functional connectivity and therefore might underlie the manifestation of neurological and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Oriane Blanquie
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
33
|
Sharp GC, Arathimos R, Reese SE, Page CM, Felix J, Küpers LK, Rifas-Shiman SL, Liu C, Burrows K, Zhao S, Magnus MC, Duijts L, Corpeleijn E, DeMeo DL, Litonjua A, Baccarelli A, Hivert MF, Oken E, Snieder H, Jaddoe V, Nystad W, London SJ, Relton CL, Zuccolo L. Maternal alcohol consumption and offspring DNA methylation: findings from six general population-based birth cohorts. Epigenomics 2017; 10:27-42. [PMID: 29172695 PMCID: PMC5753623 DOI: 10.2217/epi-2017-0095] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Alcohol consumption during pregnancy is sometimes associated with adverse outcomes in offspring, potentially mediated by epigenetic modifications. We aimed to investigate genome-wide DNA methylation in cord blood of newborns exposed to alcohol in utero. Materials & methods: We meta-analyzed information from six population-based birth cohorts within the Pregnancy and Childhood Epigenetics consortium. Results: We found no strong evidence of association at either individual CpGs or across larger regions of the genome. Conclusion: Our findings suggest no association between maternal alcohol consumption and offspring cord blood DNA methylation. This is in stark contrast to the multiple strong associations previous studies have found for maternal smoking, which is similarly socially patterned. However, it is possible that a combination of a larger sample size, higher doses, different timings of exposure, exploration of a different tissue and a more global assessment of genomic DNA methylation might show evidence of association.
Collapse
Affiliation(s)
- Gemma C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK.,School of Oral & Dental Sciences, University of Bristol, Bristol, UK
| | - Ryan Arathimos
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK
| | - Sarah E Reese
- Division of Intramural Research, Department of Health & Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Christian M Page
- Division for Mental & Physical Health, Department of Non-Communicable Diseases, Norwegian Institute of Public Health, Oslo, Norway.,Oslo Centre for Biostatistics & Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Janine Felix
- The Generation R Study Group, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Leanne K Küpers
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK.,Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sheryl L Rifas-Shiman
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Chunyu Liu
- The Framingham Heart Study, Framingham, MA, USA.,The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, & Blood Institute, Bethesda, MD, USA.,Department of Biostatistics, Boston University School of Public Health, 715 Albany St, Boston, MA, USA
| | | | - Kimberley Burrows
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK
| | - Shanshan Zhao
- Division of Intramural Research, Department of Health & Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Maria C Magnus
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK.,Division for Mental & Physical Health, Department of Non-Communicable Diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine & Allergology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Augusto Litonjua
- Channing Division of Network Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea Baccarelli
- Laboratory of Precision Environmental Biosciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA.,Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Emily Oken
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Wenche Nystad
- Division for Mental & Physical Health, Department of Non-Communicable Diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Stephanie J London
- Division of Intramural Research, Department of Health & Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK
| | - Luisa Zuccolo
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.,School of Social & Community Medicine, University of Bristol, Bristol, BS8 2BN, UK
| |
Collapse
|
34
|
Xu M, Luo J. Alcohol and Cancer Stem Cells. Cancers (Basel) 2017; 9:cancers9110158. [PMID: 29156633 PMCID: PMC5704176 DOI: 10.3390/cancers9110158] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Heavy alcohol consumption has been associated with increased risk of several cancers, including cancer of the colon, rectum, female breast, oral cavity, pharynx, larynx, liver, and esophagus. It appears that alcohol exposure not only promotes carcinogenesis but also enhances the progression and aggressiveness of existing cancers. The molecular mechanisms underlying alcohol tumor promotion, however, remain unclear. Cancer stem cells (CSC), a subpopulation of cancer cells with self-renewal and differentiation capacity, play an important role in tumor initiation, progression, metastasis, recurrence, and therapy resistance. The recent research evidence suggests that alcohol increases the CSC population in cancers, which may underlie alcohol-induced tumor promotion. This review discusses the recent progress in the research of alcohol promotion of CSC and underlying cellular/molecular mechanisms. The review will further explore the therapeutic potential of CSC inhibition in treating alcohol-induced tumor promotion.
Collapse
Affiliation(s)
- Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 1095 Veterans Drive, Lexington, KY 40536, USA.
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 1095 Veterans Drive, Lexington, KY 40536, USA.
| |
Collapse
|
35
|
Kalisch-Smith JI, Moritz KM. Detrimental effects of alcohol exposure around conception: putative mechanisms. Biochem Cell Biol 2017; 96:107-116. [PMID: 29112458 DOI: 10.1139/bcb-2017-0133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In western countries, alcohol consumption is widespread in women of reproductive age, and in binge quantities. These countries also continue to have high incidences of unplanned pregnancies, with women often reported to cease drinking after discovering their pregnancy. This suggests the early embryo may be highly exposed to the detrimental effects of alcohol during the periconception period. The periconception and pre-implantation windows, which include maturation of the oocyte, fertilisation, and morphogenesis of the pre-implantation embryo, are particularly sensitive times of development. Within the oviduct and uterus, the embryo is exposed to a unique nutritional environment to facilitate its development and establish de-novo expression of the genome through epigenetic reprogramming. Alcohol has wide-ranging effects on cellular stress, as well as hormonal, and nutrient signalling pathways, which may affect the development and metabolism of the early embryo. In this review, we summarise the adverse developmental outcomes of early exposure to alcohol (prior to implantation in animal models) and discuss the potential mechanisms for these outcomes that may occur within the protected oviductal and uterine environment. One interesting candidate is reduced retinoic acid synthesis, as it is implicated in the control of epigenetic reprogramming and cell lineage commitment, processes that have adverse consequences for the formation of the placenta, and subsequently, fetal programming.
Collapse
Affiliation(s)
- J I Kalisch-Smith
- a School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - K M Moritz
- a School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia.,b Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
| |
Collapse
|
36
|
Schang AL, Sabéran-Djoneidi D, Mezger V. The impact of epigenomic next-generation sequencing approaches on our understanding of neuropsychiatric disorders. Clin Genet 2017; 93:467-480. [DOI: 10.1111/cge.13097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/31/2022]
Affiliation(s)
- A.-L. Schang
- CNRS; UMR7216 Épigénétique et Destin Cellulaire; F-75205 Paris Cedex 13 France
- Univ Paris Diderot; Sorbonne Paris Cité, F-75205 Paris Cedex 13 France
- Département Hospitalo-Universitaire PROTECT; Paris France
| | - D. Sabéran-Djoneidi
- CNRS; UMR7216 Épigénétique et Destin Cellulaire; F-75205 Paris Cedex 13 France
- Univ Paris Diderot; Sorbonne Paris Cité, F-75205 Paris Cedex 13 France
| | - V. Mezger
- CNRS; UMR7216 Épigénétique et Destin Cellulaire; F-75205 Paris Cedex 13 France
- Univ Paris Diderot; Sorbonne Paris Cité, F-75205 Paris Cedex 13 France
| |
Collapse
|
37
|
Mandal C, Halder D, Jung KH, Chai YG. Gestational Alcohol Exposure Altered DNA Methylation Status in the Developing Fetus. Int J Mol Sci 2017; 18:ijms18071386. [PMID: 28657590 PMCID: PMC5535879 DOI: 10.3390/ijms18071386] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/27/2022] Open
Abstract
Ethanol is well known as a teratogenic factor that is capable of inducing a wide range of developmental abnormalities if the developing fetus is exposed to it. Duration and dose are the critical parameters of exposure that affect teratogenic variation to the developing fetus. It is suggested that ethanol interferes with epigenetic processes especially DNA methylation. We aimed to organize all of the available information on the alteration of DNA methylation by ethanol in utero. Thus, we have summarized all published information regarding alcohol-mediated alterations in DNA methylation during gestation. We tried to arrange information in a way that anyone can easily find the alcohol exposure time, doses, sampling time, and major changes in genomic level. Manuscript texts will also represent the correlation between ethanol metabolites and subsequent changes in methylome patterns. We hope that this review will help future researchers to further examine the issues associated with ethanol exposure.
Collapse
Affiliation(s)
- Chanchal Mandal
- Department of Molecular and Life Science, Hanyang University, 15588 Ansan, Korea.
| | - Debasish Halder
- Department of Molecular and Life Science, Hanyang University, 15588 Ansan, Korea.
| | - Kyoung Hwa Jung
- Department of Molecular and Life Science, Hanyang University, 15588 Ansan, Korea.
- Institute of Natural Science and Technology, Hanyang University, 15588 Ansan, Korea.
| | - Young Gyu Chai
- Department of Molecular and Life Science, Hanyang University, 15588 Ansan, Korea.
- Department of Bionanotechnology, Hanyang University, 04763 Seoul, Korea.
| |
Collapse
|
38
|
Abstract
Alcohol has always been present in human life, and currently it is estimated that 50% of women of childbearing age consume alcohol. It has become increasingly clear over the last years that alcohol exposure during fetal development can have detrimental effects on various organ systems, and these effects are exerted by alcohol through multiple means, including effects on free radical formation, cellular apoptosis, as well as gene expression. Fetal alcohol exposure can lead to a spectrum of short term as well as long-term problems, with Fetal Alcohol Syndrome being on the more severe end of that spectrum. This syndrome is morbid, yet preventable, and is characterized by midfacial hypoplasia, thin upper lip, widely spaced small eyes, long smooth philtrum and inner epicanthal folds. Other findings include growth restriction as well as various neurodevelopmental abnormalities. This article is the first comprehensive review combining the molecular as well as the gross physiological and anatomical effects of alcohol exposure during pregnancy on various organ systems in the body. Our knowledge of these various mechanisms is crucial for our understanding of how alcohol exposure during fetal development can lead to its detrimental effects.
Collapse
Affiliation(s)
- Marie R Nakhoul
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, D.C, USA
| | - Karl E Seif
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, D.C, USA
| | - Natasha Haddad
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, D.C, USA
| | - Georges E Haddad
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, D.C, USA
| |
Collapse
|
39
|
Lussier AA, Weinberg J, Kobor MS. Epigenetics studies of fetal alcohol spectrum disorder: where are we now? Epigenomics 2017; 9:291-311. [PMID: 28234026 PMCID: PMC5549650 DOI: 10.2217/epi-2016-0163] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adverse in utero events can alter the development and function of numerous physiological systems, giving rise to lasting neurodevelopmental deficits. In particular, data have shown that prenatal alcohol exposure can reprogram neurobiological systems, altering developmental trajectories and resulting in increased vulnerability to adverse neurobiological, behavioral and health outcomes. Increasing evidence suggests that epigenetic mechanisms are potential mediators for the reprogramming of neurobiological systems, as they may provide a link between the genome, environmental conditions and neurodevelopmental outcomes. This review outlines the current state of epigenetic research in fetal alcohol spectrum disorder, highlighting the role of epigenetic mechanisms in the reprogramming of neurobiological systems by alcohol and as potential diagnostic tools for fetal alcohol spectrum disorder. We also present an assessment of the current limitations in studies of prenatal alcohol exposure, and highlight the future steps needed in the field.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S Kobor
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Human Early Learning Partnership, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
40
|
Wang Q, Song JW, Liu Y, Zhao XX. Involvement of Wnt pathway in ethanol-induced inhibition of mouse embryonic stem cell differentiation. Alcohol 2017; 58:13-18. [PMID: 28109343 DOI: 10.1016/j.alcohol.2016.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 12/30/2022]
Abstract
Ethanol has been reported to have toxicity on embryonic stem cells (ESCs). The present study aims to address the teratogenic effects of ethanol on the growth and cardiac differentiation of ESCs. Mouse embryonic stem D3 cells were employed. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays were used to determine cytotoxicity. Quantitative real time polymerase chain reaction (qRT-PCR) and Western blotting were used to analyze the expressions of cardiac differentiation-related and Wnt signaling factors. The beating profile of cardiomyocytes was recorded to assess cardiac differentiation. Ethanol induced growth inhibition in both undifferentiated and differentiated ESCs after 5 days of exposure. Ethanol inhibited the loss of pluripotent gene expressions including Nanog, Sox2 and Oct4. The expressions of cardiac markers, Nkx2.5, Mef2c, Tbx5, dHand, αMHC, Cx43 and troponin C1, were suppressed by ethanol treatment. Furthermore, ethanol delayed cardiac differentiation of ESCs till 11 days of differentiation. The expressions of Wnt-related regulators, β-catenin and its target cyclin D1, were downregulated by ethanol. Wnt pathway agonist wnt3a could greatly rescue ethanol-induced inhibition of cardiac differentiation and Wnt-pathway-related protein expressions. These finding suggested that ethanol suppresses mouse ESC differentiation largely by inhibiting Wnt signaling pathway.
Collapse
Affiliation(s)
- Qian Wang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 ChangHai Road, Shanghai 200433, China
| | - Jing-Wen Song
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 ChangHai Road, Shanghai 200433, China
| | - Yang Liu
- The Institute of Health Science, Shanghai JiaoTong University School of Medicine, 320 YueYang Road, Shanghai 200031, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 ChangHai Road, Shanghai 200433, China.
| |
Collapse
|
41
|
Kamat PK, Mallonee CJ, George AK, Tyagi SC, Tyagi N. Homocysteine, Alcoholism, and Its Potential Epigenetic Mechanism. Alcohol Clin Exp Res 2016; 40:2474-2481. [PMID: 27805256 PMCID: PMC5133158 DOI: 10.1111/acer.13234] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
Alcohol is the most socially accepted addictive drug. Alcohol consumption is associated with some health problems such as neurological, cognitive, behavioral deficits, cancer, heart, and liver disease. Mechanisms of alcohol-induced toxicity are presently not yet clear. One of the mechanisms underlying alcohol toxicity has to do with its interaction with amino acid homocysteine (Hcy), which has been linked with brain neurotoxicity. Elevated Hcy impairs with various physiological mechanisms in the body, especially metabolic pathways. Hcy metabolism is predominantly controlled by epigenetic regulation such as DNA methylation, histone modifications, and acetylation. An alteration in these processes leads to epigenetic modification. Therefore, in this review, we summarize the role of Hcy metabolism abnormalities in alcohol-induced toxicity with epigenetic adaptation and their influences on cerebrovascular pathology.
Collapse
Affiliation(s)
- Pradip K Kamat
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida
| | - Carissa J Mallonee
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Akash K George
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
42
|
Oni EN, Hart RP. Bioinformatic Analysis of DNA Methylation in Neural Progenitor Cell Models of Alcohol Abuse. ACTA ACUST UNITED AC 2016; 2:203-210. [PMID: 27774408 DOI: 10.1007/s40495-016-0065-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several recent publications sought to investigate the effects of ethanol treatment on models of central nervous system development, specifically through changes in DNA methylation. Regulation of DNA methylation causes a long-lasting, epigenetic change in the capacity of the genome to respond to developmental or metabolic stimuli. Changes in technologies for quantifying DNA methylation have increased the ability to identify and interpret potential effects of ethanol. Here, we review these recent studies in order to evaluate the detection technologies and bioinformatic analyses. Our evaluation finds that whole- or targeted-genome sequencing combined with bisulfite conversion of unmethylated G to U residues is now the standard for assessing genome-wide effects, and specific differentially methylated regions can be validated by one of several widely-available techniques. The acceptance of these technologies should help understand how ethanol leads to life-long developmental or behavioral deficits, and, perhaps, suggest therapies to reverse these effects.
Collapse
Affiliation(s)
- Eileen N Oni
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Ronald P Hart
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
43
|
Zhang H, Gelernter J. Review: DNA methylation and alcohol use disorders: Progress and challenges. Am J Addict 2016; 26:502-515. [PMID: 27759945 DOI: 10.1111/ajad.12465] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/17/2016] [Accepted: 10/02/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Risk for alcohol use disorders (AUDs) is influenced by gene-environment interactions. Environmental factors can affect gene expression through epigenetic mechanisms such as DNA methylation. This review outlines the findings regarding the association of DNA methylation and AUDs. METHODS We searched PubMed (by April 2016) and identified 29 studies that examined the association of DNA methylation and AUDs. We also evaluated the methods used in these studies. RESULTS Two studies demonstrated elevated global (repetitive element) DNA methylation levels in AUD subjects. Fifteen candidate gene studies showed hypermethylation of promoter regions of six genes (AVP, DNMT3B, HERP, HTR3A, OPRM1, and SNCA) or hypomethylation of the GDAP1 promoter region in AUD subjects. Five genome-wide DNA methylation studies demonstrated widespread DNA methylation changes across the genome in AUD subjects. Six studies showed significant correlations of DNA methylation with gene expression in AUD subjects. Three studies revealed interactive effects of genetic variation and DNA methylation on susceptibility to AUDs. Most studies analyzed AUD-associated DNA methylation changes in the peripheral blood; a few studies examined DNA methylation changes in postmortem brains of AUD subjects. DISCUSSION AND CONCLUSIONS Chronic alcohol consumption may result in DNA methylation changes, leading to neuroadaptations that may underlie some of the mechanisms of AUD risk and persistence. Future studies are needed to confirm the few existing results, and then to elucidate whether DNA methylation changes are the cause or consequence of AUDs. SCIENTIFIC SIGNIFICANCE DNA methylation profiles may be used to assess AUD status or monitor AUD treatment response. (Am J Addict 2017;26:502-515).
Collapse
Affiliation(s)
- Huiping Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut.,Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.,Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Kim YY, Roubal I, Lee YS, Kim JS, Hoang M, Mathiyakom N, Kim Y. Alcohol-Induced Molecular Dysregulation in Human Embryonic Stem Cell-Derived Neural Precursor Cells. PLoS One 2016; 11:e0163812. [PMID: 27682028 PMCID: PMC5040434 DOI: 10.1371/journal.pone.0163812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/14/2016] [Indexed: 01/08/2023] Open
Abstract
Adverse effect of alcohol on neural function has been well documented. Especially, the teratogenic effect of alcohol on neurodevelopment during embryogenesis has been demonstrated in various models, which could be a pathologic basis for fetal alcohol spectrum disorders (FASDs). While the developmental defects from alcohol abuse during gestation have been described, the specific mechanisms by which alcohol mediates these injuries have yet to be determined. Recent studies have shown that alcohol has significant effect on molecular and cellular regulatory mechanisms in embryonic stem cell (ESC) differentiation including genes involved in neural development. To test our hypothesis that alcohol induces molecular alterations during neural differentiation we have derived neural precursor cells from pluripotent human ESCs in the presence or absence of ethanol treatment. Genome-wide transcriptomic profiling identified molecular alterations induced by ethanol exposure during neural differentiation of hESCs into neural rosettes and neural precursor cell populations. The Database for Annotation, Visualization and Integrated Discovery (DAVID) functional analysis on significantly altered genes showed potential ethanol’s effect on JAK-STAT signaling pathway, neuroactive ligand-receptor interaction, Toll-like receptor (TLR) signaling pathway, cytokine-cytokine receptor interaction and regulation of autophagy. We have further quantitatively verified ethanol-induced alterations of selected candidate genes. Among verified genes we further examined the expression of P2RX3, which is associated with nociception, a peripheral pain response. We found ethanol significantly reduced the level of P2RX3 in undifferentiated hESCs, but induced the level of P2RX3 mRNA and protein in hESC-derived NPCs. Our result suggests ethanol-induced dysregulation of P2RX3 along with alterations in molecules involved in neural activity such as neuroactive ligand-receptor interaction may be a molecular event associated with alcohol-related peripheral neuropathy of an enhanced nociceptive response.
Collapse
Affiliation(s)
- Yi Young Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Ivan Roubal
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Youn Soo Lee
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Jin Seok Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Michael Hoang
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Nathan Mathiyakom
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Yong Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
- Center for Oral and Head/Neck Oncology Research Center, Division of Oral Biology & Medicine, UCLA School of Dentistry, 10833 Le Conte Avenue, 73–022 CHS, Los Angeles, CA, 90095, United States of America
- UCLA’s Jonsson Comprehensive Cancer Center, 8–684 Factor Building, Box 951781, Los Angeles, CA, 90095, United States of America
- UCLA Broad Stem Cell Research Center, Box 957357, Los Angeles, CA, 90095, United States of America
- * E-mail:
| |
Collapse
|
45
|
Rezvanfar MA, Hodjat M, Abdollahi M. Growing knowledge of using embryonic stem cells as a novel tool in developmental risk assessment of environmental toxicants. Life Sci 2016; 158:137-60. [DOI: 10.1016/j.lfs.2016.05.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022]
|
46
|
Anstee QM, Seth D, Day CP. Genetic Factors That Affect Risk of Alcoholic and Nonalcoholic Fatty Liver Disease. Gastroenterology 2016; 150:1728-1744.e7. [PMID: 26873399 DOI: 10.1053/j.gastro.2016.01.037] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/17/2016] [Accepted: 01/20/2016] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies and candidate gene studies have informed our understanding of factors contributing to the well-recognized interindividual variation in the progression and outcomes of alcoholic liver disease and nonalcoholic fatty liver disease. We discuss the mounting evidence for shared modifiers and common pathophysiological processes that contribute to development of both diseases. We discuss the functions of proteins encoded by risk variants of genes including patatin-like phospholipase domain-containing 3 and transmembrane 6 superfamily member 2, as well as epigenetic factors that contribute to the pathogenesis of alcoholic liver disease and nonalcoholic fatty liver disease. We also discuss important areas of future genetic research and their potential to affect clinical management of patients.
Collapse
Affiliation(s)
- Quentin M Anstee
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom.
| | - Devanshi Seth
- Centenary Institute of Cancer Medicine, Royal Prince Alfred Hospital, Camperdown, Australia; Drug Health Services, Royal Prince Alfred Hospital, Camperdown, Australia; Central Clinical School, The University of Sydney, Camperdown, Australia
| | - Christopher P Day
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
47
|
Alcohol-induced suppression of KDM6B dysregulates the mineralization potential in dental pulp stem cells. Stem Cell Res 2016; 17:111-21. [PMID: 27286573 DOI: 10.1016/j.scr.2016.05.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 12/22/2022] Open
Abstract
Epigenetic changes, such as alteration of DNA methylation patterns, have been proposed as a molecular mechanism underlying the effect of alcohol on the maintenance of adult stem cells. We have performed genome-wide gene expression microarray and DNA methylome analysis to identify molecular alterations via DNA methylation changes associated with exposure of human dental pulp stem cells (DPSCs) to ethanol (EtOH). By combined analysis of the gene expression and DNA methylation, we have found a significant number of genes that are potentially regulated by EtOH-induced DNA methylation. As a focused approach, we have also performed a pathway-focused RT-PCR array analysis to examine potential molecular effects of EtOH on genes involved in epigenetic chromatin modification enzymes, fibroblastic markers, and stress and toxicity pathways in DPSCs. We have identified and verified that lysine specific demethylase 6B (KDM6B) was significantly dysregulated in DPSCs upon EtOH exposure. EtOH treatment during odontogenic/osteogenic differentiation of DPSCs suppressed the induction of KDM6B with alterations in the expression of differentiation markers. Knockdown of KDM6B resulted in a marked decrease in mineralization from implanted DPSCs in vivo. Furthermore, an ectopic expression of KDM6B in EtOH-treated DPSCs restored the expression of differentiation-related genes. Our study has demonstrated that EtOH-induced inhibition of KDM6B plays a role in the dysregulation of odontogenic/osteogenic differentiation in the DPSC model. This suggests a potential molecular mechanism for cellular insults of heavy alcohol consumption that can lead to decreased mineral deposition potentially associated with abnormalities in dental development and also osteopenia/osteoporosis, hallmark features of fetal alcohol spectrum disorders.
Collapse
|
48
|
Epigenetic Mechanisms in Developmental Alcohol-Induced Neurobehavioral Deficits. Brain Sci 2016; 6:brainsci6020012. [PMID: 27070644 PMCID: PMC4931489 DOI: 10.3390/brainsci6020012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022] Open
Abstract
Alcohol consumption during pregnancy and its damaging consequences on the developing infant brain are significant public health, social, and economic issues. The major distinctive features of prenatal alcohol exposure in humans are cognitive and behavioral dysfunction due to damage to the central nervous system (CNS), which results in a continuum of disarray that is collectively called fetal alcohol spectrum disorder (FASD). Many rodent models have been developed to understand the mechanisms of and to reproduce the human FASD phenotypes. These animal FASD studies have provided several molecular pathways that are likely responsible for the neurobehavioral abnormalities that are associated with prenatal alcohol exposure of the developing CNS. Recently, many laboratories have identified several immediate, as well as long-lasting, epigenetic modifications of DNA methylation, DNA-associated histone proteins and microRNA (miRNA) biogenesis by using a variety of epigenetic approaches in rodent FASD models. Because DNA methylation patterns, DNA-associated histone protein modifications and miRNA-regulated gene expression are crucial for synaptic plasticity and learning and memory, they can therefore offer an answer to many of the neurobehavioral abnormalities that are found in FASD. In this review, we briefly discuss the current literature of DNA methylation, DNA-associated histone proteins modification and miRNA and review recent developments concerning epigenetic changes in FASD.
Collapse
|
49
|
Immune-Mediated Inflammation May Contribute to the Pathogenesis of Cardiovascular Disease in Mucopolysaccharidosis Type I. PLoS One 2016; 11:e0150850. [PMID: 26986213 PMCID: PMC4795702 DOI: 10.1371/journal.pone.0150850] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/19/2016] [Indexed: 01/12/2023] Open
Abstract
Background Cardiovascular disease, a progressive manifestation of α-L-iduronidase deficiency or mucopolysaccharidosis type I, continues in patients both untreated and treated with hematopoietic stem cell transplantation or intravenous enzyme replacement. Few studies have examined the effects of α-L-iduronidase deficiency and subsequent glycosaminoglycan storage upon arterial gene expression to understand the pathogenesis of cardiovascular disease. Methods Gene expression in carotid artery, ascending, and descending aortas from four non-tolerized, non-enzyme treated 19 month-old mucopolysaccharidosis type I dogs was compared with expression in corresponding vascular segments from three normal, age-matched dogs. Data were analyzed using R and whole genome network correlation analysis, a bias-free method of categorizing expression level and significance into discrete modules. Genes were further categorized based on module-trait relationships. Expression of clusterin, a protein implicated in other etiologies of cardiovascular disease, was assessed in canine and murine mucopolysaccharidosis type I aortas via Western blot and in situ immunohistochemistry. Results Gene families with more than two-fold, significant increased expression involved lysosomal function, proteasome function, and immune regulation. Significantly downregulated genes were related to cellular adhesion, cytoskeletal elements, and calcium regulation. Clusterin gene overexpression (9-fold) and protein overexpression (1.3 to 1.62-fold) was confirmed and located specifically in arterial plaques of mucopolysaccharidosis-affected dogs and mice. Conclusions Overexpression of lysosomal and proteasomal-related genes are expected responses to cellular stress induced by lysosomal storage in mucopolysaccharidosis type I. Upregulation of immunity-related genes implicates the potential involvement of glycosaminoglycan-induced inflammation in the pathogenesis of mucopolysaccharidosis-related arterial disease, for which clusterin represents a potential biomarker.
Collapse
|
50
|
Thakur S, Kaur J. Regulation at multiple levels control the expression of folate transporters in liver cells in conditions of ethanol exposure and folate deficiency. Biofactors 2015; 41:232-41. [PMID: 26154406 DOI: 10.1002/biof.1217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 05/27/2015] [Indexed: 01/01/2023]
Abstract
Complex regulatory mechanisms control the expression of folate transporters within cells. Liver is the primary reserve of the folate stores within the body. As excessive alcohol consumption or inefficient dietary folate intake are known to create folate deficiency, so therefore the current study was designed to explore various regulatory mechanisms controlling the expression of folate transport in liver cells in conditions of ethanol exposure and folate deficiency. In order to see whether the effects mediated by the treatments are reversible or not, ethanol removal, and folate repletion was done after ethanol exposure and folate deficiency treatment respectively. Folate deficiency resulted an increase, whereas ethanol treatment decreased the folic acid uptake within the cells. The alterations in folic acid uptake were in agreement with the observed changes in the expression of folate transporters. Ethanol exposure resulted an increase in promoter methylation of reduced folate carrier; however, folate deficiency had no effect. The effects produced by ethanol exposure and folate deficiency were found to be reversible in nature as depicted in case of ethanol removal and folate repletion group. Rate of synthesis of folate transporters was found to be increased whereas half lives of mRNA of folate transporters was found to be decreased on folate deficiency treatment and reverse was the case on ethanol treatment. Overall, alteration in the expression of folate transporters under ethanol exposure and folate deficient conditions can be attributed to those regulatory mechanisms which work at the mRNA level.
Collapse
Affiliation(s)
- Shilpa Thakur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|