Abstract
Environmental research and public health in the 21st century face serious challenges such as increased air pollution and global warming, widespread use of potentially harmful chemicals including pesticides, plasticizers, and other endocrine disruptors, and radical changes in nutrition and lifestyle typical of modern societies. In particular, exposure to environmental and occupational toxicants may contribute to the occurrence of adverse birth outcomes, neurodevelopmental deficits, and increased risk of cancer and other multifactorial diseases such as diabetes and asthma. Rapidly evolving methodologies of exposure assessment and the conceptual framework of the Exposome, first introduced in 2005, are new frontiers of environmental research. Metabolomics and adductomics provide remarkable opportunities for a better understanding of exposure and prediction of potential adverse health outcomes. Metabolomics, the study of metabolism at the whole-body level, involves assessment of the total repertoire of small molecules present in a biological sample, shedding light on interactions between gene expression, protein expression and the environment. Advances in genomics, transcriptomics and epigenomics are generating multidimensional structures of biomarkers of effect and susceptibility, increasingly important for the understanding of molecular mechanisms and the emergence of personalized medicine. Epigenetic mechanisms, particularly DNA methylation and miRNA expression, attract increasing attention as potential links between the genetic and environmental determinants of health and disease. Unlike genetics, epigenetic mechanisms could be reversible and an understanding of their role may lead to better protection of susceptible populations and improved public health.
Keywords: Metabolomics, miRNA expression, environmental health, phthalates, DNA methylation, personalized medicine
1. Emerging Risk Factors and Public Health
The greatest public health accomplishments of the 20th century, according to the Report of the Centers for Disease Control (CDC), include healthier mothers and children due to a decrease in maternal mortality during child birth, better control of infectious diseases, and increased access to immunizations and family planning (1–2). Tobacco has been recognized as a health hazard, and significant strides have been made to protect against smoke exposure in public places. Other accomplishments ranged from motor vehicle and workplace safety; declines in deaths from heart attack and stroke; safer and healthier food and fluoridation of drinking water. Despite this progress, environmental research and public health in the 21st century face serious challenges including increased air pollution and global warming, widespread use of potentially harmful chemicals including pesticides, plasticizers, and other endocrine disruptors, and radical changes in nutrition and lifestyle typical of modern societies (3).
Many factors of environmental pollution, such as air and water pollution, as well as industrial pollution, are especially prevalent in rapidly developing economies such as China, India and some countries of Eastern Europe, Africa and South America (3). However, the impact of this pollution spreads beyond the local, and is now recognized as a driver of global climate change. Environmental and occupational exposures to toxicants may contribute to the occurrence of adverse birth outcomes, neurodevelopmental deficits, and increased risks of cancer and other common health conditions such as cardiovascular diseases, asthma, diabetes and obesity (4). These risks are not distributed evenly among populations and countries around the world, and can be modified by age, genetic makeup, SES and other factors (5–6).
2. Children are at Higher Risk
Children are more susceptible to environmental factors than adults, and fetal exposures can be especially detrimental (4). Children eat, drink and breathe more per unit of body weight than adults, and they explore their environment orally with extensive hand-to-mouth behavior (7). Moreover, newborns and young children have much lower levels of some enzymes than adults (8). For example, the paraoxonase (PON1) enzyme, initially named for its ability to hydrolyze organophosphate esters, cholinesterase inhibiting compounds often used in pesticides, is found at significantly lower levels in young children than in adults (9). Importantly, PON1 enzyme activities and genotypes have been associated with oxidative stress-related health conditions, including neurodegenerative disorders, diabetes, cardiovascular diseases, and obesity (10–13). A number of studies suggest a relationship between some PON1 polymorphisms and low PON1 protein levels with adverse development and cognition in children (14–16).
Although genetics strongly influence PON1 expression, it is clear that other factors such as epigenetics may be involved in control of PON1 enzyme variability. It is feasible that low developmental expression of the PON1 gene could be controlled by epigenetic mechanisms. The promoter polymorphism, PON1−108, was strongly associated with methylation, particularly for CpG sites located near the CpG island (17). Causal mediation analysis demonstrated statistically significant indirect effects of methylation, providing evidence that DNA methylation mediates the relationship between PON1−108 genotype and PON1 expression. These findings show that integration of genetic, epigenetic, and expression data can shed light on the functional mechanisms involving genetic and epigenetic regulation of candidate susceptibility genes like PON1. It also shows the effective application of a novel systems biology approach that relies on integration of the multidimensional data obtained by omics methodologies.
3. New Research Technologies and Environmental Health
Rapid evolution of molecular biology and genomics at the second half of the 20th century created a strong foundation for an explosion of various omic methodologies, as well as novel approaches to exposure characterization and understanding molecular mechanisms leading to human disease in the 21st century. Following the completion of the Human Genome Project in April 2003 (18), a pursuit of more powerful methodologies of sequencing the entire genome resulted in improvements to all steps of the sequencing pipeline, from bench work to bioinformatics. With a highly competitive market, innovative sequencing hardware and services advanced rapidly with increasing accuracy and decreasing costs. Sanger sequencing, or the so-called “First Generation Sequencing” that played a critical role in the completion of Human Genome Project, was an important start for the currently available techniques. The Second Generation Sequencing platforms, such as Roche 454, Illumina (MiSeq and HiSeq) and ABI SOLiD, require Polymerase Chain Reaction (PCR) in their pipeline (19). These platforms significantly improved the throughput, read length and quality of the sequencing hardware from their initial products. Recently, Illumina released the HiSeq 4000, further increasing throughput and lowering the cost per Gb of sequencing data in comparison with its competitors. The Second-Generation Sequencing platforms raised the data output from 84 kb in 2005 (first generation) to 1.8 terabases of data in a single sequencing run (Illumina 4000). HiSeqXTM Ten, released in 2014, can sequence over 45 human genomes in a single day, and new technologies promise to sequence 1000 or more genomes a day (20).
Current efforts by the genetics community to catalog genetic data worldwide have resulted in sharing data through repositories such as HapMap and the 1000 Genomes Project. These initiatives allow researchers and clinicians to determine the relative frequencies of variants in their samples compared to reference populations. For example, a GWAS study of asthma published in 2010 described location, allele frequency and local haplotype structure of approximately 15 million SNPs, 1 million short insertions and deletions and 20,000 structural variants using a whole genome sequence strategy (21). Nowadays, with existing high-throughput sequencing methodologies, an amazing amount of data can be generated. This scope of information comes with challenges in developing the best methods specific for filtering and ranking genetic variants, as well as handling the analysis of rare variants for disease and gene-by-environment (GxE) analysis.
4. Exposomics and Metabolomics
The concept of the Exposome was first introduced by Chris Wild in 2005 and has since received a lot of attention as a new approach for integral comprehensive characterization of exposure to a wide variety of nutritional, lifestyle, environmental chemicals and biological toxins (22–26). A person’s exposome is the sum total of the many exposure factors that fill the days, months, and decades of that person’s lifetime including exposures to chemicals, radiation, heat/cold, noise, food, stress, and other environmental agents; their health behaviors and activities; and the unique profile of their microbiome. Instead of characterizing exposures “one by one”, or less commonly by combined exposure to 2–3 chemicals at once, the goal is to simultaneously account for numerous factors ranging from chemical to nutritional, behavioral and environmental. As stated by Rappaport et al 2012, “Given the poor state of knowledge about health-impairing environmental exposures, epidemiologists continue to pursue narrow hypotheses that largely skirt disease etiology in favor of known environmental risk factors, even when the attributable risks are small. Although such hypothesis-driven studies confirm some environmental sources of disease, they offer only fragments to our understanding of the major causes and mechanisms of chronic diseases (27).” Methodologies for more complex assessment of total exposures are only beginning to emerge, and still have to reach the stage of development and validation enjoyed by modern genomic research.
Metabolomics has been proposed as a valuable approach to address the challenges of exposomics. Metabolomics, the study of metabolism at the whole-body level, involves assessment of the entire repertoire of small molecules present in a biological sample, shedding light on interactions between gene expression, protein expression, and the environment (28–31). Metabolomics (also known as metabonomics) allows for the full characterization of biochemical changes that occur during xenobiotic metabolism, and can therefore contribute to understanding the impact of environmental chemical exposures on human health. Recent technological developments enable comprehensive simultaneous analysis of all metabolites present in small volumes of a sample, allowing for the assessment of system-wide metabolic changes that occur as a result of an exposure or in conjunction with a health outcome (32).
Targeted metabolomics refers to methods in which specific metabolites are measured in order to characterize a pathway of interest, whereas untargeted metabolomic assays look for as many metabolites as possible on a global scale without bias (34). Transient perturbations to the transcriptome or proteome that occur in response to environmental exposures may be magnified at the level of the metabolome, making metabolomics a promising methodology for characterizing the molecular changes induced by xenobiotics and identifying new biomarkers of effect (29, 33–34). Among “omics” methodologies, metabolomics interrogates the levels of a relatively lower number of features and thus has strong statistical power compared to genome-wide and transcriptome-wide studies (35). Metabolomics is therefore a potentially sensitive method for identifying biochemical effects of external stressors. Though the developing field of “environmental metabolomics” seeks to employ metabolomic methodologies to characterize the effects of environmental exposures on organism function and health, the relationship between most of the chemicals and their effects on the human metabolome have not yet been studied (36).
For example, phthalates, endocrine disrupting chemicals that are widely used as additives in industrial and consumer products such as plasticizers, stabilizers and solubilizing agents, have been detected indoors in household air and dust, as well as in food, milk and drinking water (38). Phthalate metabolites, especially monoethyl phthalate (MEP), are commonly found in shampoos, detergents, and cosmetics (38–39). Exposure may occur through a variety of routes, including ingestion, inhalation, and dermal contact (40). Almost all U.S. residents have measurable amounts of phthalate metabolites in their urine, indicating chronic and pervasive exposure and similar exposures are reported in many other countries (41–43). Fetal exposure to phthalates is also widespread, as more than 98% of pregnant women tested have detectable levels of phthalate metabolites according to NHANES (43). Phthalates have been found in amniotic fluid, meconium, and placenta, demonstrating that phthalates cross the maternal-fetal placental barrier (43–44). Prenatal and lactational exposures have been associated with endocrine disrupting effects in animals and adverse birth outcomes in humans, strongly suggesting that early life phthalate exposures may contribute to the fetal origins of disease (41, 45–46). A number of studies have reported that levels of phthalate metabolites in the urine of pregnant women are associated with biomarkers of oxidative stress, such as isoprostane and malondialdehyde (47–48). However, limited knowledge is available on the relationship between traditional measures of phthalate exposure, characterizing up to 17 metabolites and parent compounds (49), and metabolomic markers in the same specimens as measured by targeted or untargeted methodologies.
Though phthalate metabolomic data in humans are very limited, research in animal and in vitro models suggests that metabolomic profiling can serve as a biomarker of phthalate exposure. Studies in mice and rats exposed to different dietary or prenatal doses of phthalates have demonstrated that metabolic profiles in a variety of tissues can indeed distinguish dosage groups and that profiles differ by animal sex, with male animals appearing more susceptible to metabolic dysregulation (50–52). Sumner et al. demonstrated that urinary profiles in prenatally-exposed rats could differentiate pups with or without observable reproductive effects even three weeks after exposure, indicating the potential usefulness of metabolomics as a biomarker of both exposure and effect (53).
Metabolomics research in environmentally-exposed populations may demonstrate similar effects of phthalate exposure in humans. A recent study in a Chinese male cohort used metabolomics as a tool to identify exposure biomarkers in urine. This study reported that low-level environmental phthalate exposures (DBP & MEHP) were associated with increased oxidative stress and fatty acid oxidation, and decreased prostaglandin metabolism (54). Recent discoveries using metabolic mapping technologies have helped to uncover novel pathways and metabolite-mediated posttranslational modifications, as well as their impact on physiology and disease (55).
For targeted metabolomics, a single-reaction monitoring (SRM) liquid chromatography-mass spectrometry (LC-MS/MS)-based platform, which can quantitate the levels of several hundred representative polar and nonpolar metabolites, is widely used. Other researchers use a nuclear magnetic resonance (NMR) approach (56). One technology that was developed to meet the challenge of the vast number of unknown metabolic pathways is activity-based protein profiling (ABPP) using activity-based chemical probes to assess the functional states of both characterized and uncharacterized enzymes (57–58). While targeted metabolomics is a powerful approach for quantifying changes in the levels of known metabolites in common metabolic pathways, the metabolome likely consists of many more metabolites and pathways that are yet uncharacterized. As such, untargeted and unbiased metabolomic profiling that can identify hundreds of thousand novel biomarkers and uncover unique insights into dysregulated metabolic pathways is necessary.
Adductomics is an area of research that is focused on characterizing adducts from reactions between circulating electrophiles and blood nucleophiles, and an “adductome” is defined as the totality of such adducts within a given target (27). Adducts of hemoglobin and serum albumin appear to be more informative than those of DNA and glutathione for characterizing adductomics because of their abundance and a longer half-life in human blood. So far, adductomic profiles were characterized with regard to benzene exposure, acrylamide and other environmental pollutants (59–61).
Despite exciting advances in the field of Exposomics, much more work is needed, especially with the emergence of more powerful methodologies for adductome and metabolome analysis, and cross validation with the traditional markers of exposure assessment is essential.
5. Epigenetics and Environment
Epigenetic mechanisms influence gene expression without changes in DNA sequences. Unlike genetic mutations, which lead to permanent changes of genes, epigenetic modifications are reversible and responsive to different environmental factors including lifestyle, diet and exposure to chemicals (62–63). The most widely studied epigenetic marks are DNA methylation and histone modifications. Less is known about non-coding RNAs, considered the third type of epigenetic marks (64–66). Increasing evidence has shown that epigenetic modifications, including non-coding RNA, alter or control DNA expression and the degree of DNA transcription as an adaptive response (67–69).
DNA methylation refers to the potential of a cytosine (C) base to be methylated at its 5th carbon if followed by a guanine (G) base in the DNA code, called a CpG site. The human genome contains about 30 million CpG sites distributed throughout several gene regions referred to as CpG islands, shores, shelves, and gene bodies. CpG islands are stretches of DNA with a high frequency of CpG dinucleotides that often occur in proximity to gene promoter regions (70). It was previously believed that the majority of functional changes to the methylome occurred in CpG islands; however, methylation changes along CpG shores (regions within 2kb of islands) and within the gene body may also have functional effects on gene expression (71–72). Rapidly emerging methods for measuring DNA methylation present investigators with a difficult decision in choosing the platform that provides the best balance of accuracy, coverage, reproducibility, throughput and cost (73–74).
Exploratory tools have included methylation-sensitive restriction fingerprinting, methylation-specific microarrays, and next generation sequencing. Bisulfite conversion followed by high-throughput pyrosequencing is considered the “gold standard” method for determining methylation status at specific sites (75–77). The Infinium Illumina Methylation Assay allows both genome-wide and site-specific assessment of DNA methylation. The most commonly used platform interrogates over 450,000 CpG sites (450K BeadChip) (20, 77). An even more comprehensive platform (Epic Beadchip) that covers > 850,000 CpG sites was released in 2016 (78). While Infinium BeadChips do not cover all the CpG sites in the methylome this approach can be relatively cost effective and informative in environmental pollution studies, especially when hits have been validated by other methodologies such as targeted sequencing, changes of expression and/or confirmation in the in additional cohorts (79).
DNA methylation patterns are established through critical stages of ontogenesis. The role of the prenatal period, when the embryo undergoes genome-wide DNA demethylation and remethylation, is likely to be very important in epigenome integrity and development. Environmental exposure during these sensitive time periods can result in changes, in some cases permanent, in the methylation status of DNA. The amount and patterns of DNA methylation may vary by tissue and cell type (80). Disruption of these methylation patterns has been linked to disease development and to various environmental exposures (62, 81). Some genes may become hypomethylated due to environmental exposure and others may be hypermethylated (82–85).
The rapidly expanding list of environmental exposures associated with epigenetic effects includes organochlorines, bisphenol A, persistent organic pollutants, benzene, metals, air pollution, tobacco smoke, aflatoxin B, ionizing radiation, bacteria (H.Pylori) and viruses (HPV, EBV, HBV) (86–87). Dietary and lifestyle factors such as alcohol, traumatic stress, folate deficiency, low methionine intake and aging have also been associated with epigenetic changes (82–83, 88). Limited but growing evidence also indicates that exposure to phthalates results in DNA methylation changes and may be a key mechanism by which these endocrine disruptors impact health (86). Mice exposed prenatally to di-(2-ethylhexyl) phthalate(DEHP) were seen to have increases in global DNA methylation levels and consistent increases in DNA methyltransferase protein levels, showing that in utero phthalate exposure has broad effects on DNA methylation (89). In utero exposure to DEHP was also reported to decrease androgen formation in fetal and adult rat testes by deregulating the nuclear steroid receptors through CpG hypomethylation (90). Recently, differential DNA methylation of repetitive elements, an epigenetic marker of genome instability, was reported in newborns and children with prenatal exposure to phthalate metabolites (17). These findings suggest that prenatal exposure to some endocrine disruptors may influence DNA methylation, highlighting epigenetics as a plausible biological mechanism through which environmental exposures may affect human health. When DNA methylation is analyzed in the context of population studies, it is prudent to remember that the findings will indicate associations with certain exposures, and detailed mechanistic interpretation would require additional studies in carefully designed experiments with model organisms or cell cultures (91–92).
There is a growing interest in analyzing the role of microRNAs (miRNAs) as a functionally important epigenetic mark (81, 93). MiRNAs are about 18–22 nucleotides in length and play an active role in the epigenetic regulation of gene expression in all living organisms with eukaryotic nuclear DNA (21, 94–95). The miRNAome contains more than 2,500 mature miRNAs that have been identified in humans and deposited in the miRBase (96). The majority of the miRNA target binding sites are not yet known, but putative binding sites within coding sequences can be identified by sequence complementarity to candidate miRNAs (97).
Methodologies currently available for miRNA expression analysis include Affymetrix GeneChip® miRNA 4.0 array, Nanostring and EdgeSeq miRNA whole transcriptome assay (HTG Molecular). Both Affymetrix and HTG platforms use next generation sequencing technology to quantitate miRNA expression of more than 2200 miRNAs. Compared to the arrays, the Nanostring methodology is advantageous as it is more cost effective and more appropriate for analysis of several dozens to several hundred candidate miRNAs.
MiRNAs are an excellent epigenetic biomarker to study because: 1) they are ubiquitously expressed in tissues and body fluids including blood, urine, and saliva (98–99); 2) they are released into the bloodstream from target tissues (i.e. brain, liver) and may reflect profiles of target tissue (100–101); and 3) they are highly stable and resistant to RNase activity as well as effects of pH and temperature in stored specimens over time (102). Accumulated evidence has demonstrated that most of the known miRNAs participate in normal development, as well as disease pathology, and that miRNAs are reliable biomarkers for classifying tumors and identifying tissue injury (103–108).
Although human studies of miRNA expression and environmental exposure are still limited, especially in comparison to DNA methylation, more publications have come out in recent years (87, 109–110). One study found that prenatal arsenic exposure was associated with differential miRNA expression in umbilical cord blood (111). Another study reported that among obese individuals, a conditional indirect effect of exposure to air pollution (PM10) on blood pressure was mediated by miRNA101 in people with lower BMI (112). In another study of the effects of air pollution on several candidate miRNAs, it was shown that PM10 exposure affected miRNAs that are involved in inflammatory and oxidative stress pathways (113). Exposure to black carbon was associated with differential expression of miRNA9 and miRNA96 but it was limited to only one genetic polymorphism XPO5 (114). Blood miRNAs were reported to be a sensitive indicator of environmental and occupational exposure to volatile compounds (115). It appears that the field of environmental research involving miRNA expression is developing rapidly, and these molecular biomarkers have great potential for biomonitoring, diagnostics and understanding of the mechanisms of regulation of gene-environment interactions.
Circular RNAs (also known as exonic circular RNA or circRNA) represent another exciting epigenetic mark that recently received great attention (116). It is a type of covalently closed non-colinear RNA that are linked to physiological development and various diseases (117–118). The presence of abundant circRNAs in saliva, exosomes and blood samples will make them potential diagnostic or predictive biomarkers for diseases, particularly for cancer development, progression and prognosis. Based on the current knowledge of circular RNAs, these molecules have the potential to be the "next big thing" in the omics research (119–120).
6. New Technologies in the Pipeline
A new set of sequencing technologies is rapidly replacing certain types of genomic and epigenomic microarrays as the technology of choice for quantifying and annotating different elements of the transcriptome and epigenome since they can reliably detect gene variations, gene expression and the expression of novel or known microRNAs.
For example, targeted next-generation bisulfite sequencing using library-free bisulfite padlock probes (BSPPs) will allow us to validate candidate genes and expand interrogation of CpG sites across functional gene sub-regions of interest. SNPs could be identified by using BisReadMapper software, allowing for analysis of allele specific methylation. This approach is one of the most accurate methods for bisulfite sequencing to emerge in a field undergoing rapid innovation (121). Illumina’s EPIC TruSeq Methyl Capture panel (released in 2016), an enrichment-based bisulfite sequencing method, offers another way to expand upon 850K EPIC BeadChip findings.
Given that omic research in human populations generates amazing amount of data, computationally efficient methods of integrating these datasets are becoming increasingly important (79, 122). They require a special training in bioinformatics for researchers in the environmental and public health, as well as creating an adequate, sophisticated infrastructure to meet these computational challenges.
7. Environmental Research and Personalized Medicine
Advances in genomics, transcriptomics and epigenomics are generating multidimensional structures of biomarkers of effect and susceptibility, which is increasingly important for our understanding the molecular mechanisms underlying the effects of environmental exposures and is shaping the emergence of personalized medicine (123–127). A critical aspect of understanding the role of epigenetics in gene-environment (GxE) interactions as a key mechanism of effects of environmental exposures on human health is related to differential susceptibility that may be defined by genetic make-up, age or sex. (63). Recently, the NIH Roadmap Epigenomics Consortium generated the largest collection to-date of human epigenomes for primary cells and tissues. The consortium showed that disease and trait-associated genetic variants are enriched in tissue-specific epigenomic marks, revealing biologically-relevant cell types for diverse human traits and providing a resource for understanding the molecular basis of human disease (126–128). Importantly, epigenetic marks are pliable and may be an excellent potential tool for prevention and intervention through development of new types of epigenetic drugs.
Application of novel biomedical developments, including exposomics and epigenomics, to gain a dynamic system-level and human-specific understanding of the causes of disease (128) is an important development that should enhance the contributions of toxicology and environmental research to the 21st Century paradigm in medical science and public health. This new approach can generate a more comprehensive “big picture” by linking environmental sciences with medical research through shifting the focus from animal studies and simplistic cell models to a systems biology framework (129–132).
A very promising contribution of novel omics advances is the emergence of personalized medicine. The 2015 NIH initiative on Precision Medicine is designed to “dramatically improve and encourage creative evidence based approaches” to clinical practice and disease prevention through taking advantage of molecular biology, genomics, and bioinformatics techniques (133). Currently a two-step strategy for Precision Medicine is proposed: 1. near-term focus on cancers; and 2. long-term objective to generate knowledge applicable to the whole range of health and disease. Another important aspect of the Precision Medicine Initiative is protection of sensitive sub-populations, such as pregnant women and children, from the effects of environmental exposures, drugs, poor diet, and stress. Regular comprehensive monitoring of levels of nutrients and toxicants through optimized non-invasive metabolomics or sensor-based devices already in the works by biotech companies can become a new norm in the not so distant future. Assessing differential risk will require an improved understanding of the role of genetic make-up and gene-environment interactions. This will become feasible as the efficiency and cost of these genome-wide analyses continue to improve.
In conclusion
New Age technologies and approaches can contribute to environmental research, through significantly improving biomonitoring and identification of metabolites and other chemicals, and to Precision Medicine through prevention and intervention.
Acknowledgments
This publication was made possible by grants R826886 and R82670901 from the U.S. Environmental Protection Agency (EPA) and P01 ES009605; R01ES023067 and R01ES021369 from the National Institute of Environmental Health Science (NIEHS). Its contents are solely the responsibility of the author and do not necessarily represent the official views of the NIEHS and the EPA. Kelly Nabaglo’s help with preparation of this manuscript is gratefully acknowledged.
References
- 1.American Public Health Association. Friday: Building on 20 Years of Success. doi: 10.2190/NS.20.3.l. Available at: http://www.nphw.org/tools-and-tips/themes/building-on-20-years-of-success?utm_source=NPHW%20Email&utm_medium=Email&utm_term=20%20years%20of%20success&utm_campaign=NPHW%20Friday%20Email. [DOI] [PubMed] [Google Scholar]
- 2.Centers for Disease Control and Prevention. Ten Great Public Health Achievements in the 20th Century. Available at: http://www.cdc.gov/about/history/tengpha.htm.
- 3.Smith KR, Woodward A, Campbell-Lendrum D. Health: Impacts, Adaptation, and Co-benefits. In: Field CB, Barros V, Dokken DJ, editors. Climate Change 2014: Impacts, Adaptation, and Vulnerability. Vol I: Global and Sectoral Aspects. Contribution of Working Group II to the Fifth Assessment Report of the Intergovernmental Panel on Climate Change. United Kingdom and New York: Cambridge University Press; 2014. [Google Scholar]
- 4.Landrigan PJ, Miodovnik A. Children’s Health and the Environment: An Overview. Mount Sinai J Med. 2011;78:1–10. doi: 10.1002/msj.20236. [DOI] [PubMed] [Google Scholar]
- 5.Juarez PD, Juarez PM, Hood DB, Im W, Levine RS, Kilbourne BJ, Langston MA, Al-Hamdan MZ, Crosson WL, Estes MG, Estes SM, Agboto VK, Robinson P, Wilson S, Lichtveld MY. The Public Health Exposome: A Population-Based, Exposure Science Approach to Health Disparities Research. Int J Environ Res Public Health. 2014;11:12866–12895. doi: 10.3390/ijerph111212866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Olden K, Olden HA, Lin YS. The role of the epigenome in translating neighborhood disadvantage into health disparities. Curr Environ Health Rep. 2015;2(2):163–170. doi: 10.1007/s40572-015-0048-x. [DOI] [PubMed] [Google Scholar]
- 7.National Research Council. Pesticides in the diets of infants and children. Washington D.C.: National Academy Press; 1993. [PubMed] [Google Scholar]
- 8.Padilla S, Buzzard J, Moser VC. Comparison of the role of esterases in the differential age-related sensitivity to chlorpyrifos and methamidophos. Neurotoxicology. 2000;21(1–2):49–56. [PubMed] [Google Scholar]
- 9.Huen K, Harley K, Bradman A, Eskenazi B, Holland N. Longitudinal changes in PON1 enzymatic activities in Mexican-American mothers and children with different genotypes and haplotypes. Toxicol Applied Pharmacol. 2010;244(2):181–189. doi: 10.1016/j.taap.2009.12.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bhattacharyya T, Nicholls SJ, Topol EJ, Zhang R, Yang X, Schmitt D, Shao M, Brennan DM, Ellis SG, Brennan ML, Allayee H, Lusis AJ, Hazen SL. Relationship of paraoxonase 1 (PON1) gene polymorphisms and functional activity with systemic oxidative stress and cardiovascular risk. Jama. 2008;299(11):1265–1276. doi: 10.1001/jama.299.11.1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sisk CL, Zehr JL. Pubertal hormones organize the adolescent brain and behavior. Front Neuroendocrinol. 2005;26(3–4):163–174. doi: 10.1016/j.yfrne.2005.10.003. [DOI] [PubMed] [Google Scholar]
- 12.Koncsos P, Seres I, Harangi M, Illyes I, Jozsa L, Gonczi F, Bajnok L, Paragh G. Human paraoxonase-1 activity in childhood obesity and its relation to leptin and adiponectin levels. Pediatr Res. 2009;67(3):309–313. doi: 10.1203/PDR.0b013e3181c9fb66. [DOI] [PubMed] [Google Scholar]
- 13.Costa LG, Giordano G, Furlong CE. Pharmacological and dietary modulators of paraoxonase 1 (PON1) activity and expression: the hunt goes on. Biochem Pharmacol. 2011;81(3):337–344. doi: 10.1016/j.bcp.2010.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chen J, Kumar M, Chan W, Berkowitz G, Wetmur JG. Increased influence of genetic variation on PON1 activity in neonates. Environ Health Perspect. 2003;111(11):1403–1409. doi: 10.1289/ehp.6105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Eskenazi B, Kogut K, Huen K, Harley KG, Bouchard M, Bradman A, Boyd-Barr D, Johnson C, Holland N. Organophosphate pesticide exposure, PON1, and neurodevelopment in school-age children from the CHAMACOS study. Environ Research. 2014;134:149–157. doi: 10.1016/j.envres.2014.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Engel SM, Bradman A, Wolff MS, Rauh VA, Harley KG, Yang JH, Hoepner LA, Boyd Barr D, Yolton K, Vedar MG, Xu Y, Hornung RW, Wetmur JG, Chen J, Holland N, Perera FP, Whyatt RM, Lanphear BP, Eskenazi B. Prenatal Organophosphorus Pesticide Exposure and Child Neurodevelopment at 24 Months: An Analysis of Four Birth Cohorts. Environ Health Perspect. 2015 doi: 10.1289/ehp.1409474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Huen K, Yousefi P, Street K, Eskenazi B, Holland N. PON1 as a model for integration of genetic, epigenetic, and expression data on candidate susceptibility genes. 2015 doi: 10.1093/eep/dvv003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Moraes F, Goes A. A decade of human genome project conclusion: Scientific diffusion about our genome knowledge. Biochem Mol Biol Educ. 2016;44(3):215–223. doi: 10.1002/bmb.20952. [DOI] [PubMed] [Google Scholar]
- 19.Laxman N, Rubin CJ, Mallmin H, Nilsson O, Tellgren-Roth C, Kindmark A. Second generation sequencing of microRNA in Human Bone Cells treated with Parathyroid Hormone or Dexamethasone. Bone. 2016;84:181–188. doi: 10.1016/j.bone.2015.12.053. [DOI] [PubMed] [Google Scholar]
- 20.Illumina I. An Introduction to Next-Generation Sequency Technology. Available at: http://www.illumina.com/content/dam/illumina-marketing/documents/products/illumina_sequencing_introduction.pdf. [Google Scholar]
- 21.Abecasis GR, Altshuler D, Auton A, Brooks LD, Durbin RM, Gibbs RA, Hurles ME, McVean GA. A map of human genome variation from population-scale sequencing. Nature. 2010;467(7319):1061–1073. doi: 10.1038/nature09534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wild CP. Complementing the Genome with an “Exposome”: The Outstanding Challenge of Environmental Exposure Measurement in Molecular Epidemiology. Cancer Epidemiol Biomarkers Prev. 2005;14(8):1847–1850. doi: 10.1158/1055-9965.EPI-05-0456. [DOI] [PubMed] [Google Scholar]
- 23.Wild CP. Environmental exposure measurement in cancer epidemiology. Mutagenesis. 2008;24(2):117–125. doi: 10.1093/mutage/gen061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wild CP. The exposome: from concept to utility. Int J Epidemiol. 2012:1–9. doi: 10.1093/ije/dyr236. [DOI] [PubMed] [Google Scholar]
- 25.Rappaport SM, Smith MT. Environment and disease risks. Science. 2010;330(6003):460–461. doi: 10.1126/science.1192603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Rappaport SM, Barupal DK, Wishart D, Vineis P, Scalbert A. The Blood Exposome and Its Role in Discovering Causes of Disease. Environ Health Perspect. 2014;122(8):769–774. doi: 10.1289/ehp.1308015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rappaport SM, Li H, Grigoryan H, Funk WE, Williams ER. Adductomics: Characterizing exposures to reactive electrophiles. Toxicol Lett. 2012;213:83–90. doi: 10.1016/j.toxlet.2011.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Johnson CH, Patterson AD, Idle JR, Gonzalez FJ. Xenobiotic metabolomics: major impact on the metabolome. Annu Rev Pharmacol Toxicol. 2012;52:37–56. doi: 10.1146/annurev-pharmtox-010611-134748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lankadurai BP, Nagato EG, Simpson MJ. Environmental metabolomics: an emerging approach to study organism responses to environmental stressors. Environ Rev. 2013;21(3):180–205. [Google Scholar]
- 30.Kaddurah-Daouk R, Kristal BS, Weinshilboum RM. Metabolomics: a global biochemical approach to drug response and disease. Annu Rev Pharmacol Toxicol. 2008;48:653–683. doi: 10.1146/annurev.pharmtox.48.113006.094715. [DOI] [PubMed] [Google Scholar]
- 31.Baker M. Metabolomics: from small molecules to big ideas. Nature Methods. 2010;8(2):117–121. [Google Scholar]
- 32.Patti GJ, Yanes O, Siuzdak G. Innovation: Metabolomics: the apogee of the omics trilogy. Nat Rev Mol Cell Biol. 2012;13(4):263–269. doi: 10.1038/nrm3314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.van Ravenzwaay B, Cunha GC, Leibold E, Looser R, Mellert W, Prokoudine A, Walk T, Wiemer J. The use of metabolomics for the discovery of new biomarkers of effect. Toxicol Lett. 2007;172(1–2):21–28. doi: 10.1016/j.toxlet.2007.05.021. [DOI] [PubMed] [Google Scholar]
- 34.Athersuch TJ. The role of metabolomics in characterizing the human exposome. Bioanalysis. 2012;4(18):2207–2212. doi: 10.4155/bio.12.211. [DOI] [PubMed] [Google Scholar]
- 35.van Ravenzwaay B, Herold M, Kamp H, Kapp MD, Fabian E, Looser R, Krennrich G, Mellert W, Prokoudine A, Strauss V, Walk T, Wiemer J. Metabolomics: a tool for early detection of toxicological effects and an opportunity for biology based grouping of chemicals-from QSAR to QBAR. Mutat Res. 2012;746(2):144–150. doi: 10.1016/j.mrgentox.2012.01.006. [DOI] [PubMed] [Google Scholar]
- 36.Bundy J, Davey M, Viant M. Environmental metabolomics: a critical review and future perspectives. Metabolomics. 2009;5(1):3–21. [Google Scholar]
- 37.CDC. Fourth national report on human exposure to environmental chemicals. Atlanta, GA: Centers for Disease Control and Prevention; 2009. [Google Scholar]
- 38.Silva MJ, Barr DB, Reidy JA, Malek NA, Hodge CC, Caudill SP, Brock JW, Needham LL, Calafat AM. Urinary levels of seven phthalate metabolites in the U.S. population from the national health and nutrition examination survey (NHANES) Environ Health Perspect. 2004;112:331–338. doi: 10.1289/ehp.6723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Harley K, Kogut K, Madrigal D, Cardenas M, Vera I, Meza-Alfaro G, She J, Gavin Q, Zahedi R, Bradman A, Eskenazi B, Parra K. Reducing phthalate, paraben, and phenol exposure from personal care products in adolescent girls: Findings from the hermosa intervention study. Environ Health Perspect. 2015;22 doi: 10.1289/ehp.1510514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hauser R, Calafat AM. Phthalates and human health. Occup Environ Med. 2005;62(11):806–818. doi: 10.1136/oem.2004.017590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Huang PC, Kuo PL, Chou YY, Lin SJ, Lee CC. Association between prenatal exposure to phthalates and the health of newborns. Environ Int. 2009;35(1):14–20. doi: 10.1016/j.envint.2008.05.012. [DOI] [PubMed] [Google Scholar]
- 42.Ferguson KK, Cantonwine DE, Rivera-Gonzalez LO, Loch-Caruso R, Mukherjee B, Anzalota LV, Toro D, Jimenez-Velez B, Calafat AM, Ye X, Alshawabkeh AN, Cordero JF, Meeker JD. Urinary Phthalate metabolite Associations with Biomarkers of Inflammation and Oxidative stress across pregnancy in Puerto Rico. Environ Sci Technol. 2014;48(12):7018–7025. doi: 10.1021/es502076j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Woodruff TJ, Zota AR, Schwartz JM. Environmental chemicals in pregnant women in the United States. Environ Health Perspect. 2011;119:878–885. doi: 10.1289/ehp.1002727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Swan SH. Environmental phthalate exposure in relation to reproductive outcomes and other health endpoints in humans. Environ Res. 2008;108(2):177–184. doi: 10.1016/j.envres.2008.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Parks LG, Ostby JS, Lambright CR, Abbott BD, Klinefelter GR, Barlow NJ, Gray LE., Jr The plasticizer diethylhexyl phthalate induces malformations by decreasing fetal testosterone synthesis during sexual differentiation in the male rat. Toxicol Sci. 2000;58(2):339–349. doi: 10.1093/toxsci/58.2.339. [DOI] [PubMed] [Google Scholar]
- 46.Howdeshell KL, Wilson VS, Furr J, Lambright CR, Rider CV, Blystone CR, Hotchkiss AK, Gray LE., Jr A mixture of five phthalate esters inhibits fetal testicular testosterone production in the sprague-dawley rat in a cumulative, dose-additive manner. Toxicol Sci. 2008;105(1):153–165. doi: 10.1093/toxsci/kfn077. [DOI] [PubMed] [Google Scholar]
- 47.Ferguson KK, Loch-Caruso R, Meeker JD. Exploration of oxidative stress and inflammatory markers in relation to urinary phthalate metabolites: Nhanes 1999–2006. Environ Sci Technol. 2012;46:477–485. doi: 10.1021/es202340b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Holland N, Huen K, Tran V, Street K, Nguyen B, Bradman A, Eskenazi B. Urinary phthalate metabolites and biomarkers of oxidative stress in a Mexican-American cohort: variability in early and late pregnancy. Toxics. 2016 doi: 10.3390/toxics4010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Silva MJ, Samandar E, Preau JL, Reidy JA, Needham LL, Calafat AM. Quantification of 22 phthalate metabolites in human urine. J Chromatogr B Analyt Technol Biomed Life. 2007;860:106–112. doi: 10.1016/j.jchromb.2007.10.023. [DOI] [PubMed] [Google Scholar]
- 50.Zhang J, Yan L, Tian M, Huang Q, Peng S, Dong S, Shen H. The metabonomics of combined dietary exposure to phthalates and polychlorinated biphenyls in mice. J Pharm Biomed Anal. 2012;66:287–297. doi: 10.1016/j.jpba.2012.03.045. [DOI] [PubMed] [Google Scholar]
- 51.Banerjee R, Pathmasiri W, Snyder R, McRitchie S, Sumner S. Metabolomics of brain and reproductive organs: characterizing the impact of gestational exposure to butylbenzyl phthalate on dams and resultant offspring. Metabolomics. 2012;8(6):1012–1025. [Google Scholar]
- 52.van Ravenzwaay B, Cunha GC, Strauss V, Wiemer J, Leibold E, Kamp H, Walk T, Mellert W, Looser R, Prokoudine A, Fabian E, Krennrich G, Herold M. The individual and combined metabolite profiles (metabolomics) of dibutylphthalate and di(2-ethylhexyl)phthalate following a 28-day dietary exposure in rats. Toxicol Lett. 2010;198(2):159–170. doi: 10.1016/j.toxlet.2010.06.009. [DOI] [PubMed] [Google Scholar]
- 53.Sumner S, Snyder R, Burgess J, Myers C, Tyl R, Sloan C, Fennell T. Metabolomics in the assessment of chemical-induced reproductive and developmental outcomes using non-invasive biological fluids: application to the study of butylbenzyl phthalate. J Appl Toxicol. 2009;29(8):703–714. doi: 10.1002/jat.1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang Jie, Liu L, Wang X, Huang Q, Tian M, Shen H. Low-Level Environmental Phthalate Exposure Associates with Urine Metabolome Alteration in a Chinese Male Cohort. Environ Sci Technol. 2016;50(11):5953–5960. doi: 10.1021/acs.est.6b00034. [DOI] [PubMed] [Google Scholar]
- 55.Mulvhill MM, Nomura DK. Metabolomic strategies to map functions of metabolic pathways. Am J Physiol Endocrinol Metab. 2014;307:E237–E244. doi: 10.1152/ajpendo.00228.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Larive CK, Barding GA, Dinges MM. NMR Spectroscopy for Metabolomics and Metabolic Profiling. Anal Chem. 2015;87(1):133–146. doi: 10.1021/ac504075g. [DOI] [PubMed] [Google Scholar]
- 57.Nomura DK, Dix MM, Cravatt BF. Activity-based protein profiling for biochemical pathway discovery in cancer. Nat Rev Cancer. 2010;10(9):630–638. doi: 10.1038/nrc2901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hunderdosse D, Nomura DK. Activity-based proteomic and metabolomic approaches for understanding metabolism. Curr Opinion in Biotech. 2014;28:116–126. doi: 10.1016/j.copbio.2014.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lin YS, Vermeulen R, Tsai CH, Waidyanatha S, Lan Q, Rothman N, Smith MT, Zhang L, Shen M, Li G, Yin S, Kim S, Rappaport SM. Albumin adducts of electrophilic benzene metabolites in benzene-exposed and control workers. Environ. Health Perspect. 2007;115:28–34. doi: 10.1289/ehp.8948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Obón-Santacana M, Lujan-Barroso L, Freisling H, Cadeau C, Fagherazzi G, Boutron-Ruault MC, Kaaks R, Fortner RT, Boeing H, Ramón Quirós J, Molina-Montes E, Chamosa S, Castaño JM, Ardanaz E, Khaw KT, Wareham N, Key T, Trichopoulou A, Lagiou P, Naska A, Palli D, Grioni S, Tumino R, Vineis P, De Magistris MS, Bueno-de-Mesquita HB, Peeters PH, Wennberg M, Bergdahl IA, Vesper H, Riboli E, Duell EJ. Dietary and lifestyle determinants of acrylamide and glycidamide hemoglobin adducts in non-smoking postmenopausal women from the EPIC cohort. Eur J Nutr. 2016 doi: 10.1007/s00394-016-1165-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rubino FM, Pitton M, Di Fabio D, Colombi A. Toward an “omic” physiopathology of reactive chemicals: thirty years of mass spectrometric study of the protein adducts with endogenous and xenobiotic compounds. Mass Spectrom Rev. 2009;28:725–784. doi: 10.1002/mas.20207. [DOI] [PubMed] [Google Scholar]
- 62.Foley DL, Craig JM, Morley R, Olsson CA, Dwyer T, Smith K, Saffery R. Prospects for epigenetic epidemiology. Am J Epidemiol. 2009;169(4):389–400. doi: 10.1093/aje/kwn380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Crews D, Gillette R, Miller-Crews I, Gore AC, Skinner MK. Nature, nurture and epigenetics. Mol Cell Endocrinol. 2014;398(1–2):42–52. doi: 10.1016/j.mce.2014.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Maccani MA, Marsit CJ. Epigenetics in the placenta. Am J Reprod Immunol. 2009;62(2):78–89. doi: 10.1111/j.1600-0897.2009.00716.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mccarthy MM, Nugent BM. At the frontier of epigenetics of brain sex differences. Front Behav Neurosci. 2015;9:221. doi: 10.3389/fnbeh.2015.00221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Chuang JC, Jones PA. Epigenetics and microRNAs. Pediatr Res. 2007;61(5 Pt 2):24R–29R. doi: 10.1203/pdr.0b013e3180457684. [DOI] [PubMed] [Google Scholar]
- 67.Javierre BM, Hernando H, Ballestar E. Environmental triggers and epigenetic deregulation in autoimmune disease. Discov Med. 2011;12(67):535–545. [PubMed] [Google Scholar]
- 68.Galea S, Uddin M, Koenen K. The urban environment and mental disorders: Epigenetic links. Epigenetics. 2011;6(4):400–404. doi: 10.4161/epi.6.4.14944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet. 2011;13(2):97–109. doi: 10.1038/nrg3142. [DOI] [PubMed] [Google Scholar]
- 70.Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol. 1987;196(2):261–282. doi: 10.1016/0022-2836(87)90689-9. [DOI] [PubMed] [Google Scholar]
- 71.Ball MP, Li JB, Gao Y, Lee JH, LeProust EM, Park IH, Xie B, Daley GQ, Church GM. Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat Biotechnol. 2009;27(4):361–368. doi: 10.1038/nbt.1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, Ji H, Potash J, Sabunciyan S, Feinberg AP. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–186. doi: 10.1038/ng.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Laird PW. Principles and challenges of genomewide DNA methylation analysis. Nat Rev Genet. 2010;11(3):191–203. doi: 10.1038/nrg2732. [DOI] [PubMed] [Google Scholar]
- 74.Ku CS, Naidoo N, Wu M, Soong R. Studying the epigenome using next generation sequencing. J Med Genet. 2011;48(11):721–730. doi: 10.1136/jmedgenet-2011-100242. [DOI] [PubMed] [Google Scholar]
- 75.Frommer M, McDonald Le, Millar DS, Collis CM, Watt F, Grigg GW, Molloy PL, Paul CL. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci USA. 1992;89(5):1827–1831. doi: 10.1073/pnas.89.5.1827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bock C, Tomazou EM, Brinkman AB, Muller F, Simmer F, Gu H, Jager N, Gnirke A, Stunnenberg HG, Meissner A. Quantitative comparison of genome-wide DNA methylation mapping technologies. Nat Biotechnol. 2010;28(10):1106–1114. doi: 10.1038/nbt.1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bibikova M, Barnes B, Tsan C, Ho V, Klotzle B, Le JM, Delano D, Zhang L, Schroth GP, Gunderson KL, Fan JB, Shen R. High density DNA methylation array with single CpG site resolution. Genomics. 2011;98(4):288–295. doi: 10.1016/j.ygeno.2011.07.007. [DOI] [PubMed] [Google Scholar]
- 78.Illumina Inc. Illumina. Available at: http://www.illumina.com. [Google Scholar]
- 79.Breton CV, Marsit CJ, Faustman E, Nadeau K, Goodrich JM, Dolinoy DC, Herbstman J, Holland N, LaSalle JM, Schmidt R, Yousefi P, Perera F, Joubert BR, Wiemels J, Taylor M, Yang IV, Chen R, Hew KM, Hussey Freeland DM, Miller R, Murphy S. Small magnitude effect sizes in epigenetic endpoints are important in children's environmental health studies. Environ Health Perspect. 2016 doi: 10.1289/EHP595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, Nery JR, Lee J, Ye Z, Ngo QM, Edsall L, Antosiewicz-Bourget J, Stewart R, Ruotti V, Harvey Millar A, Thomson JA, Ren B, Ecker JR. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009;462(7271):315–322. doi: 10.1038/nature08514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Marsit C. Influence of environmental exposure on human epigenetic regulation. J Experimental Biol. 2015;218:71–79. doi: 10.1242/jeb.106971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tang WY, Ho SM. Epigenetic reprogramming and imprinting in origins of disease. Rev Endocr Metab Disord. 2007;8(2):173–182. doi: 10.1007/s11154-007-9042-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Herzeg Z. Epigenetics and cancer: towards an evaluation of the impact of environmental and dietary factors. Mutagenesis. 2007;22(2):91–103. doi: 10.1093/mutage/gel068. [DOI] [PubMed] [Google Scholar]
- 84.Panni T, Mehta AJ, Schwartz JD, Baccarelli AA, Just AC, Wolf K, Wahl K, Cyrys J, Kunze S, Strauch K, Waldenberger M, Peters A. Genome-wide Analysis of DNA methylation and Fine Particulate Matter Air Pollution in Three Study Populations: KORA F3, KORA F4, and the Normative Aging Study. Environ Health Perspect. 2016;124(7):983–990. doi: 10.1289/ehp.1509966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Joubert BR, Felix JF, Yousefi P, Bakulski KM, Just AC, Breton C, et al. DNA Methylation in Newborns and Maternal Smoking in Pregnancy: Genome-wide Consortium Meta-analysis. Am J Human Genetics. 2016;98:680–696. doi: 10.1016/j.ajhg.2016.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Perera F, Herbstman J. Prenatal environmental exposures, epigenetics, and disease. Reprod Toxicol. 2011;31(3):363–373. doi: 10.1016/j.reprotox.2010.12.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Chapell G, Pogribny IP, Guyton KZ, Rusyn I. Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: A systematic literature review. Mutation Res. 2016;768:27–45. doi: 10.1016/j.mrrev.2016.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Joubert BR, den Dekker HT, Felix JF, Bohlin J, Ligthart S, et al. Maternal plasma folate impacts differential DNA methylation in an epigenome-wide meta-analysis of newborns. Nat Commun. 2015;7 doi: 10.1038/ncomms10577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wu L, et al. DNA methylation mediated by a microRNA pathway. Mol Cell. 2010;38(3):465–475. doi: 10.1016/j.molcel.2010.03.008. [DOI] [PubMed] [Google Scholar]
- 90.Martinez-Arguelles DB, Culty M, Zirkin BR, Papadopoulos V. In utero exposure to di-(2-ethylhexyl) phthalate decreases mineralocorticoid receptor expression in the adult testis. Endocrinology. 2009;150(12):5575–5585. doi: 10.1210/en.2009-0847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chadwick LH, Sawa A, Yang IV, Baccarelli A, Breakefield XO, Deng HW, Dolinoy DC, Fallin MD, Holland NT, Houseman EA, Lomvardas S, Rao M, Satterlee JS, Tyson FL, Vijayanand P, Greally JM. New insights and updated guidelines for epigenome-wide association studies. Neuroepigenetics. 2015;1:14–19. [Google Scholar]
- 92.Birney E, Smith GD, Greally JM. Epigenome-wide Association Studies and the Interpretation of Disease –Omics. PLoS One. 2016 doi: 10.1371/journal.pgen.1006105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Izzoti A, Pulliero A. The effects of environmental chemical carcinogens on the microRNA machinery. Intl J Hygiene Environ Health. 2014;217(6):601–627. doi: 10.1016/j.ijheh.2014.01.001. [DOI] [PubMed] [Google Scholar]
- 94.Postma DS, Kerkhof M, Boezen HM, Koppelman GH. Asthma and chronic obstructive pulmonary disease: common genes, common environments? Am J Respir Crit Care Med. 2011;183(12):1588–1594. doi: 10.1164/rccm.201011-1796PP. [DOI] [PubMed] [Google Scholar]
- 95.Kaneko Y, Yatagai Y, Yamada H, Iijima H, Masuko H, Sakamoto T, Hizawa N. The search for common pathways underlying asthma and COPD. Int J Chron Obstruct Pulmon Dis. 2013;8:65–78. doi: 10.2147/COPD.S39617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42(Database issue):D68–D73. doi: 10.1093/nar/gkt1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Doench JG, Sharp PA. Specificity of microRNA target selection in translational repression. Genes Dev. 2004;18(5):504–511. doi: 10.1101/gad.1184404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Landgraf P, Rusu M, Sheridan R, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129(7):1401–1414. doi: 10.1016/j.cell.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7(3) doi: 10.1371/journal.pone.0030679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zen K, Zhang CY. Circulating microRNAs: a novel class of biomarkers to diagnose and monitor human cancers. Med Res Rev. 2012;32(2):326–348. doi: 10.1002/med.20215. [DOI] [PubMed] [Google Scholar]
- 101.Grasso M, Piscopo P, Confaloni A, Denti MA. Circulating miRNAs as biomarkers for neurodegenerative disorders. Molecules. 2014;19(5):6891–6910. doi: 10.3390/molecules19056891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lussier YA, Stadler WM, Chen JL. Advantages of genomic complexity: bioinformatics opportunities in microRNA cancer signatures. J Am Med Inform Assoc. 2012;19(2):156–160. doi: 10.1136/amiajnl-2011-000419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Al-Kafaji G, Al-Mahroos G, Alsayed NA, Hasan ZA, Nawaz S, Bakhiet M. Peripheral blood microRNA-15a is a potential biomarker for type 2 diabetes mellitus and pre-diabetes. Mol Med Rep. 2015;12(5):7485–7490. doi: 10.3892/mmr.2015.4416. [DOI] [PubMed] [Google Scholar]
- 104.Dhayat SA, Husing A, Senninger N, Schmidt HH, Haier J, Wolters H, Kabar I. Circulating microRNA-200 Family as Diagnostic Marker in Hepatocellular Carcinoma. PLoS One. 2015;10(10) doi: 10.1371/journal.pone.0140066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bottoni A, Calin GA. MicroRNAs as main players in the pathogenesis of chronic lymphocytic leukemia. Microrna. 2014;2(3):158–164. doi: 10.2174/2211536602666131126002337. [DOI] [PubMed] [Google Scholar]
- 106.Seifoleslami M, Khameneie MK, Mashayekhi F, Sedaghati F, Ziari K, Mansouri K, Safari A. Identification of microRNAs (miR-203/miR-7) as potential markers for the early detection of lymph node metastases in patients with cervical cancer. Tumour Biol. 2015 doi: 10.1007/s13277-015-4265-6. [DOI] [PubMed] [Google Scholar]
- 107.Tomaszewski D. Biomarkers of Brain Damage and Postoperative Cognitive Disorders in Orthopedic Patients: An Update. Biomed Res Int. 2015;2015 doi: 10.1155/2015/402959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang L, Xu Y, Jin X, Wang Z, Wu Y, Zhao D, Chen G, Li D, Wang X, Cao H, Xie Y, Liang Z. A circulating miRNA signature as a diagnostic biomarker for non-invasive early detection of breast cancer. Breast Cancer Res Treat. 2015 doi: 10.1007/s10549-015-3591-0. [DOI] [PubMed] [Google Scholar]
- 109.Kappil M, Chen J. Environmental exposures in utero and microRNA. Curr Opin Pediatr. 2014;26(2):243–251. doi: 10.1097/MOP.0000000000000073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Vrijens K, Bollati V, Nawrot TS. MicroRNAs as Potential Signatures of Environmental Exposure or Effect: A Systematic Review. Environ Health Perspect. 2015;123(5) doi: 10.1289/ehp.1408459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Rager JE, Bailey KA, Smeester L, Miller SK, Parker JS, Laine JE, Drobná Z, Currier J, Douillet C, Olshan AF, Rubio-Andrade M, Stýblo M, García-Vargas G, Fry RC. Prenatal arsenic exposure and the epigenome: Altered microRNAs associated with innate and adaptive immune signaling in newborn cord blood. Environ Mol Mutagen. 2014;55(3):196–208. doi: 10.1002/em.21842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Motta V, Favero C, Dioni L, Iodice S, Battaglia C, Angelici L, Vigna L, Pesatori AC, Bollati V. MicroRNAs are associated with blood-pressure effects of exposure to particulate matter: Results from a mediated moderation analysis. Environ Res. 2016;146:274–281. doi: 10.1016/j.envres.2016.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Louwies T, Vuegen C, Panis LI, Cox B, Vrijens K, Nawrot T, De Boever P. miRNA expression profiles and retinal blood vessel calibers are associated with short-term particulate matter air pollution exposure. Environ Res. 2016;147:24–31. doi: 10.1016/j.envres.2016.01.027. [DOI] [PubMed] [Google Scholar]
- 114.Colicino E, Giuliano G, Power MC, Lepeule J, Wilker EH, Vokonas P, Brennan KJ, Fossati S, Hoxha M, Spiro A, 3rd, Weisskopf MG, Schwartz J, Baccarelli AA. Long-term exposure to black carbon, cognition and single nucleotide polymorphisms in microRNA processing genes in older men. Environ Intl. 2016;88:86–93. doi: 10.1016/j.envint.2015.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Song MK, Ryu JC. Blood miRNAs as sensitive and specific biological indicators of environmental and occupational exposure to volatile organic compound (VOC) Intl J Hygiene Environ Health. 2015;218(7):590–602. doi: 10.1016/j.ijheh.2015.06.002. [DOI] [PubMed] [Google Scholar]
- 116.Qu S, Yang X, Li X, Wang J, Gao Y, Shang R, Sun W, Dou K, Li H. Circular RNA: A new star of noncoding RNAs. Cancer Lett. 2015;365(2):141–148. doi: 10.1016/j.canlet.2015.06.003. [DOI] [PubMed] [Google Scholar]
- 117.Lyu D, Huang S. The emerging role and clinical implication of human exonic circular RNA. RNA Biol. 2016 doi: 10.1080/15476286.2016.1227904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Abu N, Jamal R. Circular RNAs as Promising Biomarkers: A Mini-Review. Front Physiol. 2016;7:355. doi: 10.3389/fphys.2016.00355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Floris G, Zhang L, Follesa P, Sun T. Regulatory Role of Circular RNAs and Neurological Disorders. Mol Neurobiol. 2016 doi: 10.1007/s12035-016-0055-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wang F, Nazarali AJ, Ji S. Circular RNAs as potential biomarkers for cancer diagnosis and therapy. Am J Cancer Res. 2016;6(6):1167–1176. [PMC free article] [PubMed] [Google Scholar]
- 121.Diep D, Plongthongkum N, Gore A, Fung HL, Shoemaker R, Zhang K. Library-free methylation sequencing with bisulfite padlock probes. Nat Methods. 2012;9(3):270–272. doi: 10.1038/nmeth.1871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Atwood TK, Bongcam-Rudloff E, Brazas ME, Corpas M, Gaudet P, Lewitter F, Mulder N, Palagi PM, Schneider MV, van Gelder CWG. GOBLET: The Global Organisation for Bioinformatics Learning, Education and Training. PLoS One. 2015 doi: 10.1371/journal.pcbi.1004143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, Chen R, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148(6):1293–1307. doi: 10.1016/j.cell.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Langley G, Austin CP, Balapure AK, Birnbaum LS, Bucher JR, Fentem J, et al. Lessons from Toxicology: Developing a 21st-Century Paradigm for Medical Research. Environ Health Perspect. 2015;123(11):A268–A272. doi: 10.1289/ehp.1510345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sun K, Jiang P, Chan A, Wong J, Cheng YK, Liang RH, et al. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl Acad Sci USA. 2015;112(40):E5503–E5512. doi: 10.1073/pnas.1508736112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Mullard A. The Roadmap Epigenomics Project opens new drug development avenues. Nat Rev Drug Discov. 2015;14(4):223–225. doi: 10.1038/nrd4582. [DOI] [PubMed] [Google Scholar]
- 127.Romanoski CE, Glass CK, Stunnenberg HG, Wilson L, Almouzni G. Epigenomics: Roadmap for regulation. Nature. 2015;518(7539):314–316. doi: 10.1038/518314a. [DOI] [PubMed] [Google Scholar]
- 128.Skipper M, Eccleston A, Gray N, Heemels T, Le Bot N, Marte B, Weiss U. Presenting the epigenome roadmap. Nature. 2015;518(7539):313. doi: 10.1038/518313a. [DOI] [PubMed] [Google Scholar]
- 129.National Research Council (NRC) Testing Toxicity in the 21st Century: A Vision and a Strategy. Available at: http://dels.nas.edu/Report/Toxicity-Testing-Twenty-first/11970. [Google Scholar]
- 130.Collins FS. Reeingineering translational science: the time is right. Science transl med. 2011;3(90):90cm17. doi: 10.1126/scitranslmed.3002747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Andersen BE, Naujokas MF, Suk WA. Interweaving Knowledge Resources to Address Complex Environmental Health Challenges. Environ Health Perspect. 2015;123(11):1095–1099. doi: 10.1289/ehp.1409525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ruiz P, Perlina A, Mumtaz M, Fowler BA. A system approach reveals converging molecular mechanisms that link different POPs to common metabolic diseases. Environ Health Perspect. 2016;124(7):1034–1041. doi: 10.1289/ehp.1510308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372(2):793–795. doi: 10.1056/NEJMp1500523. [DOI] [PMC free article] [PubMed] [Google Scholar]