1
|
Borrego-Ruiz A, Borrego JJ. Epigenetic Mechanisms in Aging: Extrinsic Factors and Gut Microbiome. Genes (Basel) 2024; 15:1599. [PMID: 39766866 PMCID: PMC11675900 DOI: 10.3390/genes15121599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Aging is a natural physiological process involving biological and genetic pathways. Growing evidence suggests that alterations in the epigenome during aging result in transcriptional changes, which play a significant role in the onset of age-related diseases, including cancer, cardiovascular disease, diabetes, and neurodegenerative disorders. For this reason, the epigenetic alterations in aging and age-related diseases have been reviewed, and the major extrinsic factors influencing these epigenetic alterations have been identified. In addition, the role of the gut microbiome and its metabolites as epigenetic modifiers has been addressed. RESULTS Long-term exposure to extrinsic factors such as air pollution, diet, drug use, environmental chemicals, microbial infections, physical activity, radiation, and stress provoke epigenetic changes in the host through several endocrine and immune pathways, potentially accelerating the aging process. Diverse studies have reported that the gut microbiome plays a critical role in regulating brain cell functions through DNA methylation and histone modifications. The interaction between genes and the gut microbiome serves as a source of adaptive variation, contributing to phenotypic plasticity. However, the molecular mechanisms and signaling pathways driving this process are still not fully understood. CONCLUSIONS Extrinsic factors are potential inducers of epigenetic alterations, which may have important implications for longevity. The gut microbiome serves as an epigenetic effector influencing host gene expression through histone and DNA modifications, while bidirectional interactions with the host and the underexplored roles of microbial metabolites and non-bacterial microorganisms such as fungi and viruses highlight the need for further research.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
2
|
Sampson MM, Morgan RK, Sloan SA, Bakulski KM. Single-cell investigation of lead toxicity from neurodevelopment to neurodegeneration: Current review and future opportunities. CURRENT OPINION IN TOXICOLOGY 2024; 38:100464. [PMID: 39086983 PMCID: PMC11290315 DOI: 10.1016/j.cotox.2024.100464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Human exposure to the metal lead (Pb) is prevalent and associated with adverse neurodevelopmental and neurodegenerative outcomes. Pb disrupts normal brain function by inducing oxidative stress and neuroinflammation, altering cellular metabolism, and displacing essential metals. Prior studies on the molecular impacts of Pb have examined bulk tissues, which collapse information across all cell types, or in targeted cells, which are limited to cell autonomous effects. These approaches are unable to represent the complete biological implications of Pb exposure because the brain is a cooperative network of highly heterogeneous cells, with cellular diversity and proportions shifting throughout development, by brain region, and with disease. New technologies are necessary to investigate whether Pb and other environmental exposures alter cell composition in the brain and whether they cause molecular changes in a cell-type-specific manner. Cutting-edge, single-cell approaches now enable research resolving cell-type-specific effects from bulk tissues. This article reviews existing Pb neurotoxicology studies with genome-wide molecular signatures and provides a path forward for the field to implement single-cell approaches with practical recommendations.
Collapse
Affiliation(s)
- Maureen M Sampson
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Rachel K Morgan
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Sloan
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Yu G, Wu L, Su Q, Ji X, Zhou J, Wu S, Tang Y, Li H. Neurotoxic effects of heavy metal pollutants in the environment: Focusing on epigenetic mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123563. [PMID: 38355086 DOI: 10.1016/j.envpol.2024.123563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/04/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
The pollution of heavy metals (HMs) in the environment is a significant global environmental issue, characterized by its extensive distribution, severe contamination, and profound ecological impacts. Excessive exposure to heavy metal pollutants can damage the nervous system. However, the mechanisms underlying the neurotoxicity of most heavy metals are not completely understood. Epigenetics is defined as a heritable change in gene function that can influence gene and subsequent protein expression levels without altering the DNA sequence. Growing evidence indicates that heavy metals can induce neurotoxic effects by triggering epigenetic changes and disrupting the epigenome. Compared with genetic changes, epigenetic alterations are more easily reversible. Epigenetic reprogramming techniques, drugs, and certain nutrients targeting specific epigenetic mechanisms involved in gene expression regulation are emerging as potential preventive or therapeutic tools for diseases. Therefore, this review provides a comprehensive overview of epigenetic modifications encompassing DNA/RNA methylation, histone modifications, and non-coding RNAs in the nervous system, elucidating their association with various heavy metal exposures. These primarily include manganese (Mn), mercury (Hg), lead (Pb), cobalt (Co), cadmium (Cd), nickel (Ni), sliver (Ag), toxic metalloids arsenic (As), and etc. The potential epigenetic mechanisms in the etiology, precision prevention, and target therapy of various neurodevelopmental disorders or different neurodegenerative diseases are emphasized. In addition, the current gaps in research and future areas of study are discussed. From a perspective on epigenetics, this review offers novel insights for prevention and treatment of neurotoxicity induced by heavy metal pollutants.
Collapse
Affiliation(s)
- Guangxia Yu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Lingyan Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qianqian Su
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xianqi Ji
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jinfu Zhou
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Maternity and Child Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
| | - Siying Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Ying Tang
- Fujian Center for Prevention and Control Occupational Diseases and Chemical Poisoning, Fuzhou 350125, China
| | - Huangyuan Li
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
4
|
Bjørklund G, Tippairote T, Hangan T, Chirumbolo S, Peana M. Early-Life Lead Exposure: Risks and Neurotoxic Consequences. Curr Med Chem 2024; 31:1620-1633. [PMID: 37031386 DOI: 10.2174/0929867330666230409135310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Lead (Pb) does not have any biological function in a human, and it is likely no safe level of Pb in the human body. The Pb exposure impacts are a global concern for their potential neurotoxic consequences. Despite decreasing both the environmental Pb levels and the average blood Pb levels in the survey populations, the lifetime redistribution from the tissues-stored Pb still poses neurotoxic risks from the low-level exposure in later life. The growing fetus and children hold their innate high-susceptible to these Pb-induced neurodevelopmental and neurobehavioral effects. OBJECTIVE This article aims to evaluate cumulative studies and insights on the topic of Pb neurotoxicology while assessing the emerging trends in the field. RESULTS The Pb-induced neurochemical and neuro-immunological mechanisms are likely responsible for the high-level Pb exposure with the neurodevelopmental and neurobehavioral impacts at the initial stages. Early-life Pb exposure can still produce neurodegenerative consequences in later life due to the altered epigenetic imprints and the ongoing endogenous Pb exposure. Several mechanisms contribute to the Pb-induced neurotoxic impacts, including the direct neurochemical effects, the induction of oxidative stress and inflammation through immunologic activations, and epigenetic alterations. Furthermore, the individual nutritional status, such as macro-, micro-, or antioxidant nutrients, can significantly influence the neurotoxic impacts even at low-level exposure to Pb. CONCLUSION The prevention of early-life Pb exposure is, therefore, the critical determinant for alleviating various Pb-induced neurotoxic impacts across the different age groups.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, Mo i Rana, 8610, Norway
| | - Torsak Tippairote
- Department of Nutritional and Environmental Medicine, HP Medical Center, Bangkok 10540, Thailand
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, 900470, Romania
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
- CONEM Scientific Secretary, Strada Le Grazie 9, 37134, Verona, Italy
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, Sassari, 07100, Italy
| |
Collapse
|
5
|
Mir FA, Amanullah A, Jain BP, Hyderi Z, Gautam A. Neuroepigenetics of ageing and neurodegeneration-associated dementia: An updated review. Ageing Res Rev 2023; 91:102067. [PMID: 37689143 DOI: 10.1016/j.arr.2023.102067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Gene expression is tremendously altered in the brain during memory acquisition, recall, and forgetfulness. However, non-genetic factors, including environmental elements, epigenetic changes, and lifestyle, have grabbed significant attention in recent years regarding the etiology of neurodegenerative diseases (NDD) and age-associated dementia. Epigenetic modifications are essential in regulating gene expression in all living organisms in a DNA sequence-independent manner. The genes implicated in ageing and NDD-related memory disorders are epigenetically regulated by processes such as DNA methylation, histone acetylation as well as messenger RNA editing machinery. The physiological and optimal state of the epigenome, especially within the CNS of humans, plays an intricate role in helping us adjust to the changing environment, and alterations in it cause many brain disorders, but the mechanisms behind it still need to be well understood. When fully understood, these epigenetic landscapes could act as vital targets for pharmacogenetic rescue strategies for treating several diseases, including neurodegeneration- and age-induced dementia. Keeping this objective in mind, this updated review summarises the epigenetic changes associated with age and neurodegeneration-associated dementia.
Collapse
Affiliation(s)
- Fayaz Ahmad Mir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Zeeshan Hyderi
- Department of Biotechnology, Alagappa University, Karaikudi, India
| | - Akash Gautam
- Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, India.
| |
Collapse
|
6
|
Bolognesi G, Bacalini MG, Pirazzini C, Garagnani P, Giuliani C. Evolutionary Implications of Environmental Toxicant Exposure. Biomedicines 2022; 10:3090. [PMID: 36551846 PMCID: PMC9775150 DOI: 10.3390/biomedicines10123090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Homo sapiens have been exposed to various toxins and harmful compounds that change according to various phases of human evolution. Population genetics studies showed that such exposures lead to adaptive genetic changes; while observing present exposures to different toxicants, the first molecular mechanism that confers plasticity is epigenetic remodeling and, in particular, DNA methylation variation, a molecular mechanism proposed for medium-term adaptation. A large amount of scientific literature from clinical and medical studies revealed the high impact of such exposure on human biology; thus, in this review, we examine and infer the impact that different environmental toxicants may have in shaping human evolution. We first describe how environmental toxicants shape natural human variation in terms of genetic and epigenetic diversity, and then we describe how DNA methylation may influence mutation rate and, thus, genetic variability. We describe the impact of these substances on biological fitness in terms of reproduction and survival, and in conclusion, we focus on their effect on brain evolution and physiology.
Collapse
Affiliation(s)
- Giorgia Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, via San Giacomo 12, 40126 Bologna, Italy
- Laboratory of Molecular Anthropology, Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, via Francesco Selmi 3, 40126 Bologna, Italy
| | - Maria Giulia Bacalini
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, via Altura 3, 40139 Bologna, Italy
| | - Chiara Pirazzini
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, via Altura 3, 40139 Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, via San Giacomo 12, 40126 Bologna, Italy
| | - Cristina Giuliani
- Laboratory of Molecular Anthropology, Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, via Francesco Selmi 3, 40126 Bologna, Italy
| |
Collapse
|
7
|
Liu M, Liu R, Wang R, Ba Y, Yu F, Deng Q, Huang H. Lead-induced neurodevelopmental lesion and epigenetic landscape: Implication in neurological disorders. J Appl Toxicol 2022. [PMID: 36433892 DOI: 10.1002/jat.4419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/20/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Lead (Pb) was implicated in multiple genotoxic, neuroepigenotoxic, and chromosomal-toxic mechanisms and interacted with varying synaptic plasticity pathways, likely underpinning previous reports of links between Pb and cognitive impairment. Epigenetic changes have emerged as a promising biomarker for neurological disorders, including cognitive disorders, Alzheimer's disease (AD), and Parkinson's disease (PD). In the present review, special attention is paid to neural epigenetic features and mechanisms that can alter gene expression patterns upon environmental Pb exposure in rodents, primates, and zebrafish. Epigenetic modifications have also been discussed in population studies and cell experiment. Further, we explore growing evidence of potential linkage between Pb-induced disruption of regulatory pathway and neurodevelopmental and neurological disorders both in vivo and in vitro. These findings uncover how epigenome in neurons facilitates the development and function of the brain in response to Pb insult.
Collapse
Affiliation(s)
- Mengchen Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Rundong Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Ruike Wang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Yue Ba
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Fangfang Yu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Qihong Deng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| |
Collapse
|
8
|
Tasin FR, Ahmed A, Halder D, Mandal C. On-going consequences of in utero exposure of Pb: An epigenetic perspective. J Appl Toxicol 2022; 42:1553-1569. [PMID: 35023172 DOI: 10.1002/jat.4287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 11/08/2022]
Abstract
Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hyper- and hypo-methylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration- and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as - impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.
Collapse
Affiliation(s)
- Fahim Rejanur Tasin
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Asif Ahmed
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Debasish Halder
- Rare Disease research center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
9
|
Sánchez OF, Lin LF, Xie J, Freeman JL, Yuan C. Lead exposure induces dysregulation of constitutive heterochromatin hallmarks in live cells. Curr Res Toxicol 2021; 3:100061. [PMID: 35005634 PMCID: PMC8717252 DOI: 10.1016/j.crtox.2021.12.001] [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] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/28/2022] Open
Abstract
Lead (Pb) is a heavy metal contaminant commonly found in air, soil, and drinking water due to legacy uses. Excretion of ingested Pb can result in extensive kidney damages due to elevated oxidative stress. Epigenetic alterations induced by exposure to Pb have also been implied but remain poorly understood. In this work, we assessed changes in repressive epigenetic marks, namely DNA methylation (meCpG) and histone 3 lysine 9 tri-methylation (H3K9me3) after exposure to Pb. Live cell epigenetic probes coupled to bimolecular fluorescence complementation (BiFC) were used to monitor changes in the selected epigenetic marks. Exposure to Pb significantly lowered meCpG and H3K9me3 levels in HEK293T cells suggesting global changes in constitutive heterochromatin. A heterodimeric pair of probes that tags chromatin regions enriched in both meCpG and H3K9me3 further confirmed our findings. The observed epigenetic changes can be partially attributed to aberrant transcriptional changes induced by Pb, such as overexpression of TET1 after Pb exposure. Lastly, we monitored changes in selected heterochromatin marks after removal of Pb and found that changes in these markers do not immediately recover to their original level suggesting potential long-term damages to chromatin structure.
Collapse
Affiliation(s)
- Oscar F. Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Li F. Lin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Jennifer L. Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Center of Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Purdue Center of Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
10
|
Mäkinen H, van Oers K, Eeva T, Ruuskanen S. The effect of experimental lead pollution on DNA methylation in a wild bird population. Epigenetics 2021; 17:625-641. [PMID: 34369261 DOI: 10.1080/15592294.2021.1943863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Anthropogenic pollution is known to negatively influence an organism's physiology, behaviour, and fitness. Epigenetic regulation, such as DNA methylation, has been hypothesized as a potential mechanism to mediate such effects, yet studies in wild species are lacking. We first investigated the effects of early-life exposure to the heavy metal lead (Pb) on DNA methylation levels in a wild population of great tits (Parus major), by experimentally exposing nestlings to Pb at environmentally relevant levels. Secondly, we compared nestling DNA methylation from a population exposed to long-term heavy metal pollution (close to a copper smelter), where birds suffer from pollution-related decrease in food quality, and a control population. For both comparisons, the analysis of about one million CpGs covering most of the annotated genes revealed that pollution-related changes in DNA methylation were not genome wide, but enriched for genes underlying developmental processes. However, the results were not consistent when using binomial or beta binomial regression highlighting the difficulty of modelling variance in CpGs. Our study indicates that post-natal anthropogenic heavy metal exposure can affect methylation levels of development related genes in a wild bird population.
Collapse
Affiliation(s)
- Hannu Mäkinen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Turku, Finland
| | - Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands
| | - Tapio Eeva
- Department of Biological and Environmental Sciences, University of Jyväskylä, Turku, Finland
| | - Suvi Ruuskanen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Turku, Finland
| |
Collapse
|
11
|
Migliore L, Nicolì V, Stoccoro A. Gender Specific Differences in Disease Susceptibility: The Role of Epigenetics. Biomedicines 2021; 9:652. [PMID: 34200989 PMCID: PMC8228628 DOI: 10.3390/biomedicines9060652] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 01/08/2023] Open
Abstract
Many complex traits or diseases, such as infectious and autoimmune diseases, cancer, xenobiotics exposure, neurodevelopmental and neurodegenerative diseases, as well as the outcome of vaccination, show a differential susceptibility between males and females. In general, the female immune system responds more efficiently to pathogens. However, this can lead to over-reactive immune responses, which may explain the higher presence of autoimmune diseases in women, but also potentially the more adverse effects of vaccination in females compared with in males. Many clinical and epidemiological studies reported, for the SARS-CoV-2 infection, a gender-biased differential response; however, the majority of reports dealt with a comparable morbidity, with males, however, showing higher COVID-19 adverse outcomes. Although gender differences in immune responses have been studied predominantly within the context of sex hormone effects, some other mechanisms have been invoked: cellular mosaicism, skewed X chromosome inactivation, genes escaping X chromosome inactivation, and miRNAs encoded on the X chromosome. The hormonal hypothesis as well as other mechanisms will be examined and discussed in the light of the most recent epigenetic findings in the field, as the concept that epigenetics is the unifying mechanism in explaining gender-specific differences is increasingly emerging.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
- Department of Laboratory Medicine, Azienda Ospedaliero Universitaria Pisana, 56124 Pisa, Italy
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
| | - Andrea Stoccoro
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
| |
Collapse
|
12
|
Hernández-Coro A, Sánchez-Hernández BE, Montes S, Martínez-Lazcano JC, González-Guevara E, Pérez-Severiano F. Alterations in gene expression due to chronic lead exposure induce behavioral changes. Neurosci Biobehav Rev 2021; 126:361-367. [PMID: 33819547 DOI: 10.1016/j.neubiorev.2021.03.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/15/2020] [Accepted: 03/27/2021] [Indexed: 02/01/2023]
Abstract
Lead (Pb) is a pollutant commonly found in the environment, despite the implementation of public health policies intended to remove it. Due to its chemical characteristics as a divalent ion, Pb interacts with cells, enzymes, and tissues, causing pathological, physical, and behavioral alterations. Recent biotechnological advances have helped us to understand the mechanisms underlying the damage caused by Pb in human populations and in experimental models, and new evidence on the epigenetic alterations caused by exposition to environmental Pb is available. It is known that Pb exposure impacts on behavior (causing aggressiveness, anxiety, and depression), leading to learning deficit and locomotor activity alterations, and its presence has been linked with the abnormal release of neurotransmitters and other biochemical changes involved in these disorders. Still, further reductionist studies are required to determine the effects of Pb exposure on DNA and protein expression and understand the processes underlying the diseases caused by Pb. This will also indicate possible therapeutic targets to offset the negative effects of the heavy metal. By elucidating the epigenetic changes involved, it would be possible to manipulate them and propose novel therapeutic approaches in this area. This review is aimed to provide an overview of studies that link Pb exposure to behavioral changes, as well as biochemical and epigenetic alterations at a neurotransmitter level, considering the importance of this metal in behavior abnormalities.
Collapse
Affiliation(s)
- Abraham Hernández-Coro
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, La Fama, 14269, Mexico City, Mexico
| | - Beatriz Eugenia Sánchez-Hernández
- Departamento de Genética, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga #15, Col. Belisario Domínguez Sección 16, Tlalpan, 14080, Mexico City, Mexico
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur # 3877, La Fama, 14269. Mexico City, Mexico
| | - Juan Carlos Martínez-Lazcano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, La Fama, 14269, Mexico City, Mexico
| | - Edith González-Guevara
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, La Fama, 14269, Mexico City, Mexico
| | - Francisca Pérez-Severiano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, La Fama, 14269, Mexico City, Mexico.
| |
Collapse
|
13
|
Ijomone OM, Ijomone OK, Iroegbu JD, Ifenatuoha CW, Olung NF, Aschner M. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology 2020; 81:51-65. [PMID: 32882300 PMCID: PMC7708394 DOI: 10.1016/j.neuro.2020.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Continuous globalization and industrialization have ensured metals are an increasing aspect of daily life. Their usefulness in manufacturing has made them vital to national commerce, security and global economy. However, excess exposure to metals, particularly as a result of environmental contamination or occupational exposures, has been detrimental to overall health. Excess exposure to several metals is considered environmental risk in the aetiology of several neurological and neurodegenerative diseases. Metal-induced neurotoxicity has been a major health concern globally with intensive research to unravel the mechanisms associated with it. Recently, greater focus has been directed at epigenetics to better characterize the underlying mechanisms of metal-induced neurotoxicity. Epigenetic changes are those modifications on the DNA that can turn genes on or off without altering the DNA sequence. This review discusses how epigenetic changes such as DNA methylation, post translational histone modification and noncoding RNA-mediated gene silencing mediate the neurotoxic effects of several metals, focusing on manganese, arsenic, nickel, cadmium, lead, and mercury.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
| | - Olayemi K Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Nzube F Olung
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
14
|
Wang K, Liu S, Svoboda LK, Rygiel CA, Neier K, Jones TR, Colacino JA, Dolinoy DC, Sartor MA. Tissue- and Sex-Specific DNA Methylation Changes in Mice Perinatally Exposed to Lead (Pb). Front Genet 2020; 11:840. [PMID: 32973866 PMCID: PMC7472839 DOI: 10.3389/fgene.2020.00840] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Lead (Pb) is a well-known toxicant that interferes with the development of a child’s nervous and metabolic systems and increases the risk of developing diseases later in life. Although studies have investigated epigenetic effects associated with Pb exposure, knowledge of genome-wide changes with in vivo low dose perinatal Pb exposure in multiple tissues is limited. Within the Toxicant Exposures and Responses by Genomic and Epigenomic Regulators of Transcription (TaRGET II) consortium, we utilized a mouse model to investigate tissue- and sex-specific DNA methylation. Dams were assigned to control or Pb-acetate water, respectively. Exposures started 2 weeks prior to mating and continued until weaning at post-natal day 21 (PND21). Liver and blood were collected from PND21 mice, and the DNA methylome was assessed using enhanced reduced representation bisulfite sequencing (ERRBS). We identified ∼1000 perinatal Pb exposure related differentially methylated cytosines (DMCs) for each tissue- and sex-specific comparison, and hundreds of tissue- and sex-specific differentially methylated regions (DMRs). Several mouse imprinted genes were differentially methylated across both tissues in males and females. Overall, our findings demonstrate that perinatal Pb exposure can induce tissue- and sex-specific DNA methylation changes and provide information for future Pb studies in humans.
Collapse
Affiliation(s)
- Kai Wang
- Department of Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Siyu Liu
- Department of Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Laurie K Svoboda
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Christine A Rygiel
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Kari Neier
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Tamara R Jones
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Justin A Colacino
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, MI, United States.,Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
15
|
Zare Mehrjerdi F, Niknazar S, Yadegari M, Akbari FA, Pirmoradi Z, Khaksari M. Carvacrol reduces hippocampal cell death and improves learning and memory deficits following lead-induced neurotoxicity via antioxidant activity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1229-1237. [PMID: 32303785 DOI: 10.1007/s00210-020-01866-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Carvacrol is a monoterpene with neuroprotective effects in several animal models of neurodegeneration, including epilepsy, ischemia, and traumatic neuronal events. In this study, we aimed to examine the effects of carvacrol on neurodegeneration induced by lead acetate in rats. A total of 50 male Wistar rats were divided into five equal groups. The control group received drinking water, while the neurotoxic group was exposed to 500 ppm of lead acetate in drinking water for 40 days. The three remaining groups, which were also exposed to 500 ppm of lead acetate, received carvacrol at doses of 25, 50, and 100 mg/kg orally for 40 days. The Morris water maze test was employed to examine spatial learning and memory. Pathological damage to the hippocampus was determined by Nissl staining. The level of malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) were detected using biochemical analysis and the free radical scavenging activity as evaluated by the DPPH test. Administration of carvacrol significantly restored learning and memory impairment induced by lead acetate. Moreover, carvacrol ameliorated neurodegeneration, antioxidative capacity, and lipid peroxidation in the hippocampus of rats exposed to lead. The present results provide a rationale for the inhibitory role of carvacrol in the attenuation of lead-induced neurotoxicity.
Collapse
Affiliation(s)
- Fatemeh Zare Mehrjerdi
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Somayeh Niknazar
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Yadegari
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Fatemeh Ali Akbari
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Zeynab Pirmoradi
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mehdi Khaksari
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
16
|
Kochmanski J, Bernstein AI. The Impact of Environmental Factors on 5-Hydroxymethylcytosine in the Brain. Curr Environ Health Rep 2020; 7:109-120. [PMID: 32020534 PMCID: PMC7809708 DOI: 10.1007/s40572-020-00268-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The aims of this review are to evaluate the methods used to measure 5-hydroxymethylcytosine (5-hmC), and then summarize the available data investigating the impact of environmental factors on 5-hydroxymethylcytosine (5-hmC) in the brain. RECENT FINDINGS Recent research has shown that some environmental factors, including exposure to exogenous chemicals, stress, altered diet, and exercise, are all associated with 5-hmC variation in the brain. However, due to a lack of specificity in the methods used to generate a majority of the available data, it cannot be determined whether environment-induced changes in 5-hmC occur in specific biological pathways. Environment appears to shape 5-hmC levels in the brain, but the available literature is hampered by limitations in measurement methods. The field of neuroepigenetics needs to adopt new tools to increase the specificity of its data and enhance biological interpretation of exposure-related changes in 5-hmC. This will help improve understanding of the potential roles for environmental factors and 5-hmC in neurological disease.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Department of Translational Neuroscience, Grand Rapids Research Center, Michigan State University College of Human Medicine, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Alison I Bernstein
- Department of Translational Neuroscience, Grand Rapids Research Center, Michigan State University College of Human Medicine, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
17
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
18
|
Sex-Dependent Effects of Developmental Lead Exposure in Wistar Rats: Evidence from Behavioral and Molecular Correlates. Int J Mol Sci 2020; 21:ijms21082664. [PMID: 32290408 PMCID: PMC7216048 DOI: 10.3390/ijms21082664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Lead (Pb) exposure in early life affects brain development resulting in cognitive and behavioral deficits. Epidemiologic and experimental evidence of sex as an effect modifier of developmental Pb exposure is emerging. In the present study, we investigated Pb effects on behavior and mechanisms of neuroplasticity in the hippocampus and potential sex differences. To this aim, dams were exposed, from one month pre-mating to offspring weaning, to Pb via drinking water at 5 mg/kg body weight per day. In the offspring of both sexes, the longitudinal assessment of motor, emotional, and cognitive end points was performed. We also evaluated the expression and synaptic distribution of N-methyl-D-Aspartate receptor (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits at post-natal day (pnd) 23 and 70 in the hippocampus. Neonatal motor patterns and explorative behavior in offspring were affected in both sexes. Pb effects in emotional response and memory retention were observed in adult females only, preceded by increased levels of GluN2A and GluA1 subunits at the post-synapse at pnd 23. These data suggest that Pb exposure during development affects glutamatergic receptors distribution at the post-synaptic spine in females. These effects may contribute to alterations in selected behavioral domains.
Collapse
|
19
|
Onuzulu CD, Rotimi OA, Rotimi SO. Epigenetic modifications associated with in utero exposure to endocrine disrupting chemicals BPA, DDT and Pb. REVIEWS ON ENVIRONMENTAL HEALTH 2019; 34:309-325. [PMID: 31271561 DOI: 10.1515/reveh-2018-0059] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 04/03/2019] [Indexed: 06/09/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are xenobiotics which adversely modify the hormone system. The endocrine system is most vulnerable to assaults by endocrine disruptors during the prenatal and early development window, and effects may persist into adulthood and across generations. The prenatal stage is a period of vulnerability to environmental chemicals because the epigenome is usually reprogrammed during this period. Bisphenol A (BPA), lead (Pb), and dichlorodiphenyltrichloroethane (DDT) were chosen for critical review because they have become serious public health concerns globally, especially in Africa where they are widely used without any regulation. In this review, we introduce EDCs and describe the various modes of action of EDCs and the importance of the prenatal and developmental windows to EDC exposure. We give a brief overview of epigenetics and describe the various epigenetic mechanisms: DNA methylation, histone modifications and non-coding RNAs, and how each of them affects gene expression. We then summarize findings from previous studies on the effects of prenatal exposure to the endocrine disruptors BPA, Pb and DDT on each of the previously described epigenetic mechanisms. We also discuss how the epigenetic alterations caused by these EDCs may be related to disease processes.
Collapse
Affiliation(s)
- Chinonye Doris Onuzulu
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| | - Oluwakemi Anuoluwapo Rotimi
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| | - Solomon Oladapo Rotimi
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| |
Collapse
|
20
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Dou JF, Farooqui Z, Faulk CD, Barks AK, Jones T, Dolinoy DC, Bakulski KM. Perinatal Lead (Pb) Exposure and Cortical Neuron-Specific DNA Methylation in Male Mice. Genes (Basel) 2019; 10:genes10040274. [PMID: 30987383 PMCID: PMC6523909 DOI: 10.3390/genes10040274] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/25/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Abstract
: Lead (Pb) exposure is associated with a wide range of neurological deficits. Environmental exposures may impact epigenetic changes, such as DNA methylation, and can affect neurodevelopmental outcomes over the life-course. Mating mice were obtained from a genetically invariant C57BL/6J background agouti viable yellow Avy strain. Virgin dams (a/a) were randomly assigned 0 ppm (control), 2.1 ppm (low), or 32 ppm (high) Pb-acetate water two weeks prior to mating with male mice (Avy/a), and this continued through weaning. At age 10 months, cortex neuronal nuclei were separated with NeuN⁺ antibodies in male mice to investigate neuron-specific genome-wide promoter DNA methylation using the Roche NimbleGen Mouse 3x720K CpG Island Promoter Array in nine pooled samples (three per dose). Several probes reached p-value < 10-5 , all of which were hypomethylated: 12 for high Pb (minimum false discovery rate (FDR) = 0.16, largest intensity ratio difference = -2.1) and 7 for low Pb (minimum FDR = 0.56, largest intensity ratio difference = -2.2). Consistent with previous results in bulk tissue, we observed a weak association between early-life exposure to Pb and DNA hypomethylation, with some affected genes related to neurodevelopment or cognitive function. Although these analyses were limited to males, data indicate that non-dividing cells such as neurons can be carriers of long-term epigenetic changes induced in development.
Collapse
Affiliation(s)
- John F Dou
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Zishaan Farooqui
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Christopher D Faulk
- Department of Animal Science, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, St. Paul, MN 55108, USA.
| | - Amanda K Barks
- Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55454, USA.
| | - Tamara Jones
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Zhang W, Zhang Y, Zheng Y, Zheng M, Sun N, Yang X, Gao Y. Progress in Research on Brain Development and Function of Mice During Weaning. Curr Protein Pept Sci 2019; 20:705-712. [PMID: 30678620 DOI: 10.2174/1389203720666190125095819] [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: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/13/2019] [Indexed: 01/15/2023]
Abstract
Lactation is a critical phase for brain function development. New dietary experiences of mouse caused by weaning can regulate brain development and function, increase their response to food and environment, and eventually give rise to corresponding behavioral changes. Changes in weaning time induce the alteration of brain tissues morphology and molecular characteristics, glial cell activity and behaviors in the offspring. In addition, it is also sensitive to the intervention of environment and drugs during this period. That is to say, the study focused on brain development and function based on mouse weaning is critical to demonstrate the underlying pathogenesis of neuropsychiatric diseases and find new drug targets. This article mainly focuses on the developmental differentiation of the brain during lactation, especially during weaning in mice.
Collapse
Affiliation(s)
- Wenjie Zhang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yueling Zhang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, China
| | - Yuanjia Zheng
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingxuan Zheng
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu, China
| | - Nannan Sun
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Yang
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu, China
| | - Yong Gao
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
23
|
Beyrne CC, González RM, Iusem ND. Strategy for the analysis of tissue-specific methylation changes without physical isolation. Epigenetics 2019; 14:41-51. [PMID: 30632887 DOI: 10.1080/15592294.2019.1565589] [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] [Indexed: 12/14/2022] Open
Abstract
One common experimental hurdle that arises when explore patterns of cytosine methylation is the generation of data derived from a single specific tissue, often arduous to isolate from a heterogeneous biospecimen. Here we show a new strategy for exploring environment- or mutation-caused changes in cell type- or tissue-specific methylation landscapes, which requires neither transgenic reporter cell lines nor physical separation. This approach takes advantage of a known distinct methylation signature existing in only one of the tissues within an organ under a particular condition. From the information on such compared published methylomes, one can design a set of PCR primers that specifically amplify bisulfite-converted DNA of two nearby genomic regions of interest, thus allowing for tissue-specific DNA methylation data. To validate the performance of the approach, we designed primers able to amplify a portion of a gene in the context of root biology: the Arabidopsis homeotic gene Glabra-2 (Gl2), expressed only in epidermis during cell differentiation. We found that the extent of methylated cytosines appears remarkably different when root epidermis-specific primers were used vs. non-specific ones under three genetic backgrounds involving mutations in genes also associated with the establishment of cell identity. Although the genetic or environmental perturbations to be studied might modify methylation in the primer-annealing zone, leading to a possible misinterpretation of the data, the strategy presented here can become a useful first round screening tool to detect differences in tissue-specific epigenetic status under new conditions.
Collapse
Affiliation(s)
- Cecilia C Beyrne
- a Instituto de Fisiología , Biología Molecular y Neurociencias (IFIByNE); CONICET , Buenos Aires , Argentina
| | - Rodrigo M González
- a Instituto de Fisiología , Biología Molecular y Neurociencias (IFIByNE); CONICET , Buenos Aires , Argentina
| | - Norberto D Iusem
- a Instituto de Fisiología , Biología Molecular y Neurociencias (IFIByNE); CONICET , Buenos Aires , Argentina.,b Departamento de Fisiología, Biología Molecular y Celular (FBMC); Facultad de Ciencias Exactas y Naturales , Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
24
|
Jaffee SR. Lead exposure and child maltreatment as models for how to conceptualize early-in-life risk factors for violence. Infant Ment Health J 2019; 40:23-38. [DOI: 10.1002/imhj.21756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
25
|
del Blanco B, Barco A. Impact of environmental conditions and chemicals on the neuronal epigenome. Curr Opin Chem Biol 2018; 45:157-165. [DOI: 10.1016/j.cbpa.2018.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 05/28/2018] [Accepted: 06/02/2018] [Indexed: 01/04/2023]
|
26
|
Sobolewski M, Varma G, Adams B, Anderson DW, Schneider JS, Cory-Slechta DA. Developmental Lead Exposure and Prenatal Stress Result in Sex-Specific Reprograming of Adult Stress Physiology and Epigenetic Profiles in Brain. Toxicol Sci 2018; 163:478-489. [PMID: 29481626 PMCID: PMC5974781 DOI: 10.1093/toxsci/kfy046] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Developmental exposure to lead (Pb) and prenatal stress (PS) both impair cognition, which could derive from their joint targeting of the hypothalamic-pituitary-adrenal axis and the brain mesocorticolimbic (MESO) system, including frontal cortex (FC) and hippocampus (HIPP). Glucocorticoids modulate both FC and HIPP function and associated mediation of cognitive and other behavioral functions. This study sought to determine whether developmental Pb ± PS exposures altered glucocorticoid-related epigenetic profiles in brain MESO regions in offspring of female mice exposed to 0 or 100 ppm Pb acetate drinking water from 2 mos prior to breeding until weaning, with half further exposed to prenatal restraint stress from gestational day 11-18. Overall, changes in females occured in response to Pb exposure. In males, however, Pb-induced neurotoxicity was modulated by PS. Changes in serum corticosterone levels were seen in males, while glucocorticoid receptor changes were seen in both sexes. In contrast, both Pb and PS broadly impacted brain DNA methyltransferases and binding proteins, particularly DNMT1, DNMT3a and methyl-CpG-binding protein 2, with patterns that differed by sex and brain regions. Specifically, in males, effects on FC epigenetic modifiers were primarily influenced by Pb, whereas extensive changes in HIPP were produced by PS. In females, Pb exposure and not PS primarily altered epigenetic modifiers in both FC and HIPP. Collectively, these findings indicate that epigenetic mechanisms may underlie associated neurotoxicity of Pb and of PS, particularly associated cognitive deficits. However, mechanisms by which this may occur will be different in males versus females.
Collapse
Affiliation(s)
- Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York,To whom correspondence should be addressed at Department of Environmental Medicine, University of Rochester School of Medicine, University of Rochester Medical Center, Box EHSC, Rochester, NY 14642. Fax: 585-256-2591; E-mail:
| | - Garima Varma
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Beth Adams
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - David W Anderson
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York
| |
Collapse
|
27
|
Singh G, Singh V, Wang ZX, Voisin G, Lefebvre F, Navenot JM, Evans B, Verma M, Anderson DW, Schneider JS. Effects of developmental lead exposure on the hippocampal methylome: Influences of sex and timing and level of exposure. Toxicol Lett 2018; 290:63-72. [PMID: 29571894 DOI: 10.1016/j.toxlet.2018.03.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/15/2018] [Accepted: 03/19/2018] [Indexed: 12/21/2022]
Abstract
Developmental lead (Pb) exposure results in persistent cognitive/behavioral impairments as well as an elevated risk for developing a variety of diseases in later life. Environmental exposures during development can result in a variety of epigenetic changes, including alterations in DNA methylation, that can influence gene expression patterns and affect the function and development of the nervous system. The present promoter-based methylation microarray profiling study explored the extent to which developmental Pb exposure may modify the methylome of a brain region, hippocampus, known to be sensitive to the effects of Pb exposure. Male and female Long Evans rats were exposed to 0 ppm, 150 ppm, 375 ppm, or 750 ppm Pb through perinatal exposures (gestation through lactation), early postnatal exposures (birth through weaning), or long-term postnatal exposures (birth through postnatal day 55). Results showed a significant contribution of sex to the hippocampal methylome and effects of Pb exposure level, with non-linear dose response effects on methylation. Surprisingly, the developmental period of exposure contributed only a small amount of variance to the overall data and gene ontology (GO) analysis revealed the largest number of overrepresented GO terms in the groups with the lowest level of exposure. The highest number of significant differentially methylated regions was found in females exposed to Pb at the lowest exposure level. Our data reinforce the significant effect that low level Pb exposure may have on gene-specific DNA methylation patterns in brain and that this occurs in a sex-dependent manner.
Collapse
Affiliation(s)
- G Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| | - V Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zi-Xuan Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - G Voisin
- Atelerics S.E.N.C, Montreal, QC, Canada
| | - F Lefebvre
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - J-M Navenot
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - B Evans
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - M Verma
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - D W Anderson
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
28
|
Singh G, Singh V, Sobolewski M, Cory-Slechta DA, Schneider JS. Sex-Dependent Effects of Developmental Lead Exposure on the Brain. Front Genet 2018; 9:89. [PMID: 29662502 PMCID: PMC5890196 DOI: 10.3389/fgene.2018.00089] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 11/23/2022] Open
Abstract
The role of sex as an effect modifier of developmental lead (Pb) exposure has until recently received little attention. Lead exposure in early life can affect brain development with persisting influences on cognitive and behavioral functioning, as well as, elevated risks for developing a variety of diseases and disorders in later life. Although both sexes are affected by Pb exposure, the incidence, manifestation, and severity of outcomes appears to differ in males and females. Results from epidemiologic and animal studies indicate significant effect modification by sex, however, the results are not consistent across studies. Unfortunately, only a limited number of human epidemiological studies have included both sexes in independent outcome analyses limiting our ability to draw definitive conclusions regarding sex-differentiated outcomes. Additionally, due to various methodological differences across studies, there is still not a good mechanistic understanding of the molecular effects of lead on the brain and the factors that influence differential responses to Pb based on sex. In this review, focused on prenatal and postnatal Pb exposures in humans and animal models, we discuss current literature supporting sex differences in outcomes in response to Pb exposure and explore some of the ideas regarding potential molecular mechanisms that may contribute to sex-related differences in outcomes from developmental Pb exposure. The sex-dependent variability in outcomes from developmental Pb exposure may arise from a combination of complex factors, including, but not limited to, intrinsic sex-specific molecular/genetic mechanisms and external risk factors including sex-specific responses to environmental stressors which may act through shared epigenetic pathways to influence the genome and behavioral output.
Collapse
Affiliation(s)
- Garima Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Vikrant Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Deborah A. Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Jay S. Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
29
|
McCabe C, Anderson OS, Montrose L, Neier K, Dolinoy DC. Sexually Dimorphic Effects of Early-Life Exposures to Endocrine Disruptors: Sex-Specific Epigenetic Reprogramming as a Potential Mechanism. Curr Environ Health Rep 2018; 4:426-438. [PMID: 28980159 DOI: 10.1007/s40572-017-0170-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The genetic material of every organism exists within the context of regulatory networks that govern gene expression-collectively called the epigenome. Animal models and human birth cohort studies have revealed key developmental periods that are important for epigenetic programming and vulnerable to environmental insults. Thus, epigenetics represent a potential mechanism through which sexually dimorphic effects of early-life exposures such as endocrine-disrupting chemicals (EDCs) manifest. RECENT FINDINGS Several animal studies, and to a lesser extent human studies, have evaluated life-course sexually dimorphic health effects following developmental toxicant exposures; many fewer studies, however, have evaluated epigenetics as a mechanism mediating developmental exposures and later outcomes. To evaluate epigenetic reprogramming as a mechanistic link of sexually dimorphic early-life EDCs exposures, the following criteria should be met: (1) well-characterized exposure paradigm that includes relevant windows for developmental epigenetic reprogramming; (2) evaluation of sex-specific exposure-related epigenetic change; and (3) observation of a sexually dimorphic phenotype in either childhood, adolescence, or adulthood.
Collapse
Affiliation(s)
- Carolyn McCabe
- Nutritonal Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA
| | - Olivia S Anderson
- Nutritonal Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA
| | - Luke Montrose
- Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA
| | - Kari Neier
- Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA
| | - Dana C Dolinoy
- Nutritonal Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA. .,Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI, 48109-2029, USA.
| |
Collapse
|
30
|
Montrose L, Faulk C, Francis J, Dolinoy D. Perinatal lead (Pb) exposure results in sex and tissue-dependent adult DNA methylation alterations in murine IAP transposons. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:540-550. [PMID: 28833526 PMCID: PMC5784428 DOI: 10.1002/em.22119] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/25/2017] [Accepted: 05/25/2017] [Indexed: 05/17/2023]
Abstract
Epidemiological and animal data suggest that adult chronic disease is influenced by early-life exposure-induced changes to the epigenome. Previously, we observed that perinatal lead (Pb) exposure results in persistent murine metabolic- and activity-related effects. Using phylogenetic and DNA methylation analysis, we have also identified novel intracisternal A particle (IAP) retrotransposons exhibiting regions of variable methylation as candidate loci for environmental effects on the epigenome. Here, we now evaluate brain and kidney DNA methylation profiles of four representative IAPs in adult mice exposed to human physiologically relevant levels of Pb two weeks prior to mating through lactation. When IAPs across the genome were evaluated globally, average (sd) methylation levels were 92.84% (3.74) differing by tissue (P < 0.001), but not sex or dose. By contrast, the four individual IAPs displayed tissue-specific Pb and sex effects. Medium Pb-exposed mice had 3.86% less brain methylation at IAP 110 (P < 0.01), while high Pb-exposed mice had 2.83% less brain methylation at IAP 236 (P = 0.01) and 1.77% less at IAP 506 (P = 0.05). Individual IAP DNA methylation differed by sex for IAP 110 in the brain and kidney, IAP 236 in the kidney, and IAP 1259 in the kidney. Using Tomtom, we identified three binding motifs that matched to each of our novel IAPs impacted by Pb, one of which (HMGA2) has been linked to metabolic-related conditions in both mice and humans. Thus, these recently identified IAPs display tissue-specific environmental lability as well as sex-specific differences supporting an epigenetic link between early exposure to Pb and later-in-life health outcomes. Environ. Mol. Mutagen. 58:540-550, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- L. Montrose
- Environmental Health Sciences, University of Michigan
| | - C. Faulk
- Animal Science, University of Minnesota
| | - J. Francis
- Environmental Health Sciences, University of Michigan
| | - D.C. Dolinoy
- Environmental Health Sciences, University of Michigan
- Nutritional Sciences, University of Michigan
- Corresponding author: Dana C. Dolinoy, 1415 Washington Heights, Ann Arbor, Michigan 48109-2029, Tel: 734 647-3155,
| |
Collapse
|
31
|
Cory-Slechta DA, Sobolewski M, Varma G, Schneider JS. Developmental Lead and/or Prenatal Stress Exposures Followed by Different Types of Behavioral Experience Result in the Divergence of Brain Epigenetic Profiles in a Sex, Brain Region, and Time-Dependent Manner: Implications for Neurotoxicology. CURRENT OPINION IN TOXICOLOGY 2017; 6:60-70. [PMID: 29430559 DOI: 10.1016/j.cotox.2017.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over a lifetime, early developmental exposures to neurocognitive risk factors, such as lead (Pb) exposures and prenatal stress (PS), will be followed by multiple varied behavioral experiences. Pb, PS and behavioral experience can each influence brain epigenetic profiles. Our recent studies show a greater level of complexity, however, as all three factors interact within each sex to generate differential adult variation in global post-translational histone modifications (PTHMs), which may result in fundamentally different consequences for life-long learning and behavioral function. We have reported that PTHM profiles differ by sex, brain region and time point of measurement following developmental exposures to Pb±PS, resulting in different profiles for each unique combination of these parameters. Imposing differing behavioral experience following developmental Pb±PS results in additional divergence of PTHM profiles, again in a sex, brain region and time-dependent manner, further increasing complexity. Such findings underscore the need to link highly localized and variable epigenetic changes along single genes to the highly-integrated brain functional connectome that is ultimately responsible for governing behavioral function. Here we advance the idea that increased understanding may be achieved through iterative reductionist and holistic approaches. Implications for experimental design of animal studies of developmental exposures to neurotoxicants include the necessity of a 'no behavioral experience' group, given that epigenetic changes in response to behavioral testing can confound effects of the neurotoxicant itself. They also suggest the potential utility of the inclusion of salient behavioral experiences as a potential effect modifier in epidemiological studies.
Collapse
Affiliation(s)
- Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical School, Rochester, NY
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical School, Rochester, NY
| | - G Varma
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - J S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
32
|
Varma G, Sobolewski M, Cory-Slechta DA, Schneider JS. Sex- and brain region- specific effects of prenatal stress and lead exposure on permissive and repressive post-translational histone modifications from embryonic development through adulthood. Neurotoxicology 2017; 62:207-217. [PMID: 28712943 PMCID: PMC5623619 DOI: 10.1016/j.neuro.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022]
Abstract
Developmental exposure to prenatal stress (PS) and lead (Pb) can affect brain development and adversely influence behavior and cognition. Epigenetic-based gene regulation is crucial for normal brain development and mis-regulation, in any form, can result in neurodevelopmental disorders. Post-translational histone modifications (PTHMs) are an integral and dynamic component of the epigenetic machinery involved in gene regulation. Exposures to Pb and/or PS may alter PTHM profiles, promoting lifelong alterations in brain function observed following Pb±PS exposure. Here we examined the effects of Pb±PS on global levels of activating marks H3K9Ac and H3K4Me3 and repressive marks H3K9Me2 and H3K27Me3 at different developmental stages: E18, PND0, PND6 and PND60. Dams were exposed to 0 or 100ppm Pb beginning 2 months prior to breeding followed by no PS (NS) or PS resulting in 4 offspring treatment groups per sex: 0-NS (control), 0-PS, 100-NS and 100-PS. Global levels of PTHMs varied from E18 through adulthood even in control mice, and were influenced by sex and brain-region. The developmental trajectory of these PTHM levels was further modified by Pb±PS in a sex-, brain region- and age-dependent manner. Females showed a preferential response to Pb alone in frontal cortex (FC) and differentially to PS alone and combined Pb+PS in hippocampus (HIPP). In males, PS-induced increases in PTHM levels in FC, whereas PS produced reductions in HIPP. Pb±PS-based changes in PTHM levels continued to be observed in adulthood (PND60), demonstrating the lasting effect of these early life environmental events on these histone marks. These results indicate that epigenetic consequences of Pb±PS and their contribution to mechanisms of toxicity are sex dependent. Additional studies will assist in understanding the functional significance of these changes in PTHM levels on expression of individual genes, functional pathways, and ultimately, their behavioral consequences.
Collapse
Affiliation(s)
- G Varma
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - M Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester NY, United States
| | - D A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester NY, United States
| | - J S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
33
|
Villalba-Benito L, Torroglosa A, Fernández RM, Ruíz-Ferrer M, Moya-Jiménez MJ, Antiñolo G, Borrego S. Overexpression of DNMT3b target genes during Enteric Nervous System development contribute to the onset of Hirschsprung disease. Sci Rep 2017; 7:6221. [PMID: 28740121 PMCID: PMC5524929 DOI: 10.1038/s41598-017-06539-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/09/2017] [Indexed: 12/27/2022] Open
Abstract
Hirschsprung disease (HSCR) is attributed to a failure of neural crest cells (NCCs) to migrate, proliferate, differentiate and/or survive in the bowel wall during embryonic Enteric Nervous System (ENS) development. ENS formation is the result from a specific gene expression pattern regulated by epigenetic events, such DNA methylation by the DNA methyltransferases (DNMTs), among other mechanisms. Specifically, DNMT3b de novo methyltransferase is associated with NCCs development and has been shown to be implicated in ENS formation and in HSCR. Aiming to elucidate the specific mechanism underlying the DNMT3b role in such processes, we have performed a chromatin immunoprecipitation coupled with massively parallel sequencing analysis to identify the DNMT3B target genes in enteric precursor cells (EPCs) from mice. Moreover, the expression patterns of those target genes have been analyzed in human EPCs from HSCR patients in comparison with controls. Additionally, we have carried out a search of rare variants in those genes in a HSCR series. Through this approach we found 9 genes showing a significantly different expression level in both groups. Therefore, those genes may have a role in the proper human ENS formation and a failure in their expression pattern might contribute to this pathology.
Collapse
Affiliation(s)
- Leticia Villalba-Benito
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Ana Torroglosa
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Raquel María Fernández
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Macarena Ruíz-Ferrer
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - María José Moya-Jiménez
- Department of Pediatric Surgery, University Hospital Virgen del Rocío, Seville, 41013, Spain
| | - Guillermo Antiñolo
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain
| | - Salud Borrego
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, 41013, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, 41013, Spain.
| |
Collapse
|
34
|
Khan H, Singh RD, Tiwari R, Gangopadhyay S, Roy SK, Singh D, Srivastava V. Mercury exposure induces cytoskeleton disruption and loss of renal function through epigenetic modulation of MMP9 expression. Toxicology 2017; 386:28-39. [DOI: 10.1016/j.tox.2017.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 11/30/2022]
|
35
|
Wang J, Gao ZY, Yan J, Ying XL, Tong SL, Yan CH. Sex differences in the effects of prenatal lead exposure on birth outcomes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 225:193-200. [PMID: 28371734 DOI: 10.1016/j.envpol.2017.03.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 06/07/2023]
Abstract
Studies on the associations between prenatal lead exposure and birth outcomes have been inconsistent, and few data are available on the sex differences in these associations. We measured the cord blood lead levels of newborns in Shanghai and determined their associations with birth outcomes, which included birth weight, birth length, head circumference, and the ponderal index, in the total sample and within sex subgroups. A total of 1009 mother-infant pairs were enrolled from 10 hospitals in Shanghai between September 2008 and October 2009. The geometric mean of the cord blood lead concentrations was 4.07 μg/dl (95% CI: 3.98-4.17 μg/dl). A significant inverse association was found between cord blood lead levels and head circumference only in the male subgroup, and increasing cord blood lead levels were related to significant decreases in the ponderal index only in females. The birth weights of the male infants were positively associated with cord blood lead levels; after adjusting for the maternal intake frequency of preserved eggs, the estimated mean differences in birth weights decreased by 11.7% for each 1-unit increase in the log10-transformed cord blood lead concentration. Our findings suggest that prenatal lead exposure may have sex-specific effects on birth outcomes and that maternal dietary intake may be a potential confounder in these relationships. Further studies on this topic are highly warranted.
Collapse
Affiliation(s)
- Ju Wang
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China; Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen-Yan Gao
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lan Ying
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Lu Tong
- School of Public Health and Social Work, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Chong-Huai Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Zhou S, Morozova TV, Hussain YN, Luoma SE, McCoy L, Yamamoto A, Mackay TF, Anholt RR. The Genetic Basis for Variation in Sensitivity to Lead Toxicity in Drosophila melanogaster. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1062-70. [PMID: 26859824 PMCID: PMC4937873 DOI: 10.1289/ehp.1510513] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/19/2015] [Accepted: 01/21/2016] [Indexed: 05/05/2023]
Abstract
BACKGROUND Lead toxicity presents a worldwide health problem, especially due to its adverse effects on cognitive development in children. However, identifying genes that give rise to individual variation in susceptibility to lead toxicity is challenging in human populations. OBJECTIVES Our goal was to use Drosophila melanogaster to identify evolutionarily conserved candidate genes associated with individual variation in susceptibility to lead exposure. METHODS To identify candidate genes associated with variation in susceptibility to lead toxicity, we measured effects of lead exposure on development time, viability and adult activity in the Drosophila melanogaster Genetic Reference Panel (DGRP) and performed genome-wide association analyses to identify candidate genes. We used mutants to assess functional causality of candidate genes and constructed a genetic network associated with variation in sensitivity to lead exposure, on which we could superimpose human orthologs. RESULTS We found substantial heritabilities for all three traits and identified candidate genes associated with variation in susceptibility to lead exposure for each phenotype. The genetic architectures that determine variation in sensitivity to lead exposure are highly polygenic. Gene ontology and network analyses showed enrichment of genes associated with early development and function of the nervous system. CONCLUSIONS Drosophila melanogaster presents an advantageous model to study the genetic underpinnings of variation in susceptibility to lead toxicity. Evolutionary conservation of cellular pathways that respond to toxic exposure allows predictions regarding orthologous genes and pathways across phyla. Thus, studies in the D. melanogaster model system can identify candidate susceptibility genes to guide subsequent studies in human populations. CITATION Zhou S, Morozova TV, Hussain YN, Luoma SE, McCoy L, Yamamoto A, Mackay TF, Anholt RR. 2016. The genetic basis for variation in sensitivity to lead toxicity in Drosophila melanogaster. Environ Health Perspect 124:1062-1070; http://dx.doi.org/10.1289/ehp.1510513.
Collapse
Affiliation(s)
- Shanshan Zhou
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Tatiana V. Morozova
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Yasmeen N. Hussain
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Department of Biochemistry and Physiology, School of Bioscience and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Sarah E. Luoma
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Lenovia McCoy
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Akihiko Yamamoto
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Trudy F.C. Mackay
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Robert R.H. Anholt
- W.M. Keck Center for Behavioral Biology, Program in Genetics, and
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Address correspondence to R.R.H. Anholt, W.M. Keck Center for Behavioral Biology, 3510 Thomas Hall, North Carolina State University, Campus Box 7614, Raleigh, NC 27695-7614 USA. Telephone: (919) 515-1173. E-mail:
| |
Collapse
|
37
|
Keil KP, Lein PJ. DNA methylation: a mechanism linking environmental chemical exposures to risk of autism spectrum disorders? ENVIRONMENTAL EPIGENETICS 2016; 2:dvv012. [PMID: 27158529 PMCID: PMC4856164 DOI: 10.1093/eep/dvv012] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
There is now compelling evidence that gene by environment interactions are important in the etiology of autism spectrum disorders (ASDs). However, the mechanisms by which environmental factors interact with genetic susceptibilities to confer individual risk for ASD remain a significant knowledge gap in the field. The epigenome, and in particular DNA methylation, is a critical gene expression regulatory mechanism in normal and pathogenic brain development. DNA methylation can be influenced by environmental factors such as diet, hormones, stress, drugs, or exposure to environmental chemicals, suggesting that environmental factors may contribute to adverse neurodevelopmental outcomes of relevance to ASD via effects on DNA methylation in the developing brain. In this review, we describe epidemiological and experimental evidence implicating altered DNA methylation as a potential mechanism by which environmental chemicals confer risk for ASD, using polychlorinated biphenyls (PCBs), lead, and bisphenol A (BPA) as examples. Understanding how environmental chemical exposures influence DNA methylation and how these epigenetic changes modulate the risk and/or severity of ASD will not only provide mechanistic insight regarding gene-environment interactions of relevance to ASD but may also suggest potential intervention strategies for these and potentially other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kimberly P. Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
- *Correspondence address. Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA. Tel:
(530) 752-1970
; Fax:
(530) 752-7690
; E-mail:
| |
Collapse
|
38
|
Lindquist DM, Beckwith T, Cecil KM, Sánchez-Martín FJ, Landero-Figueroa J, Puga A. Prenatal and early postnatal lead exposure in mice: neuroimaging findings. Quant Imaging Med Surg 2015; 5:511-8. [PMID: 26435914 DOI: 10.3978/j.issn.2223-4292.2015.07.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Childhood lead exposure has been linked to adult gray matter loss accompanied by changes in myelination and neurochemistry noninvasively revealed by magnetic resonance imaging (MRI) methods. However, the extent, duration and timing of lead exposure required to produce such imaging changes in humans are difficult to ascertain. METHODS To determine if such changes are related to early exposure to low levels of lead, we treated mouse dams with 0, 3, or 30 ppm of lead acetate in drinking water for 2 months prior to mating through gestation until weaning of the offspring at post-natal day 21. Two male and two female pups from each litter were imaged at post-natal day 60. Volumetric, diffusion tensor imaging and magnetic resonance spectroscopy (MRS) measurements were obtained using a seven Tesla Bruker animal MRI scanner. RESULTS Postnatal blood lead levels were identical between groups at the time of imaging. No effects of lead exposure were detected in the volumetric or MRS data. Mean diffusivity in the hippocampus showed significant effects of lead exposure and gender. CONCLUSIONS These data suggest that low-level, gestational lead exposure in a mouse model produces minimal changes observed by MRI.
Collapse
Affiliation(s)
- Diana M Lindquist
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Travis Beckwith
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kim M Cecil
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Francisco Javier Sánchez-Martín
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Julio Landero-Figueroa
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Alvaro Puga
- 1 Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA ; 2 Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA ; 3 Metallomics Center of the Americas, Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|