Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 25.
Published in final edited form as: Rev Environ Health. 2019 Sep 25;34(3):251–259. doi: 10.1515/reveh-2019-0030

Application of metabolomics to characterize environmental pollutant toxicity and disease risks

Pan Deng 1,2, Xusheng Li 1,3, Michael C Petriello 1,4, Chunyan Wang 1,2, Andrew J Morris 1,4, Bernhard Hennig 1,2
PMCID: PMC6915040  NIHMSID: NIHMS1062296  PMID: 31408434

Abstract

The increased incidence of non-communicable human diseases may be attributed, at least partially, to exposures to toxic chemicals such as persistent organic pollutants, air pollutants, and heavy metals. Given the high mortality and morbidity of pollutant exposure associated diseases, a better understanding of related mechanisms of toxicity and impacts on endogenous host metabolism is needed. The metabolome represents the collection of the intermediates and end products of cellular processes, and is the most proximal reporter of the body’s response to environmental exposures and pathological processes. Metabolomics is a powerful tool for studying how organisms interact with their environment and how these interactions shape diseases related to pollutant exposure. This mini review discusses potential biological mechanisms that link pollutant exposure to metabolic disturbances and chronic human diseases, with a focus on recent studies that demonstrate the application of metabolomics as a tool to elucidate biochemical modes of actions of various environmental pollutants. In addition, classes of metabolites that have been shown to be modulated by multiple environmental pollutants will be discussed with an emphasis on their use as potential early biomarkers of disease risks. Taken together, metabolomics is a useful and versatile tool for characterizing the disease risks and mechanisms associated with various environmental pollutants.

Keywords: metabolomics, pollutant, disease, toxicity, POP, heavy metal

Introduction

Most non-communicable disease risk is determined by both genetic and a broad range of environmental factors. In addition to lifestyle-dependent factors, such as diet, exercise and other behavioral choices, exposure to toxic environmental chemicals is one example of environmental factors that can modify disease outcome [13]. The pathology of diseases such as cardiovascular, neurodegenerative, immunological, respiratory disorders and cancers can be modified or accelerated due to exposure to toxic pro-inflammatory chemicals. Other lifestyle and environmental factors that can contribute to these diseases include diet, tobacco smoking, exposures to radiation, pathogens, allergens and psychological stress [4]. Although genetic factors are a significant determinant of the risk of these chronic diseases, exposure to toxic environmental chemicals is recognized as a major independent risk factor [57]. As industrialization increases worldwide, it is expected that exposure risks may increase which has become a global health challenge. The World Health Organization (WHO) estimates that environmental exposures accounted for 12.6 million deaths worldwide and 22% of the global burden of disease in 2012 [8].

Metabolomics is the field of science that characterizes endogenous and exogenous metabolites within a cell, tissue, or biofluid of an organism in response to external stressors such as disease, contaminant exposure, or nutritional imbalances [9]. Nuclear magnetic resonance (NMR) and mass spectrometry (MS) are the two major analytical tools that are routinely used to obtain metabolomics data sets [10]. Metabolomic analyses can be categorized as either non-targeted or targeted with non-targeted metabolomics detecting as many distinct features as possible in a single analysis. In contrast, targeted metabolomics analyzes a specific group, class, or species of metabolites and is especially useful for absolute quantitation. As the pathways and networks of metabolites have been extensively studied (Figure 1), changes in metabolites can simultaneously reveal variations in enzymatic levels, activities and/or gene expression patterns.

Figure 1. Systematic workflow of metabolomic studies.

Figure 1.

Biological samples from cells, animal and/or human bio-fluids or tissues are extracted for NMR or MS data acquisition. Bioinformatic analysis, or data processing, including feature finding, metabolite identification and pathway analysis, can then be used to identify changes in cellular metabolism, and ultimately elucidate the biochemical/molecular mechanisms underpinning disease pathogenesis.

Metabolomics allows researchers to assess the cellular response to external stimuli and can help to improve the understanding of mode-of-action of a given compound [11]. Metabolomic analysis of genetically modified animal models is a powerful tool for understanding downstream ramifications of targeted gene dysfunction [1214]. For example, Ahr-knockout male mice were used in a metabolomic study of 2,3,7,8-tetrachlorodibenzofuran (TCDF), and it was found that ingestion of TCDF altered hepatic lipogenesis, gluconeogenesis, and glycogenolysis in an AHR-dependent manner [14]. Additionally, animal disease models were used in metabolomic studies to investigate mechanisms of pollutant related pathology process. For example, a Helicobacter-infected mouse model was used to study the role of gut microbiome perturbation in arsenic (As) induced disease. The results suggested that gut microbiome perturbation can exacerbate metabolic disorders induced by As exposure[15]. It should be noted that although gender differences in tissue distribution and toxicity have been reported for many environmental pollutants [1618], there are few studies exploring pollutants-induced metabolome changes in female animal models. The application of metabolomic technologies in epidemiological studies is a growing research area with the potential to better characterize human exposures, detect early markers of disease, understand disease etiology, improve diagnosis of disease, and track disease progression [19]. Table 1, and described in the following sections of this mini-review, provides an overview of metabolomic-based epidemiological studies for some pollutant classes. “Meet-in-the-middle” (MITM) approaches involve a prospective search for intermediate biomarkers, which are elevated in subjects who eventually develop disease and a retrospective search for links of such biomarkers to past environmental exposures [20]. This approach has been conceived as a strategy to identify biomarkers that are not only related to specific exposures but also predictive of disease outcome, and it is gaining popularity for the ability to reveal important linkages along the exposure-outcome pathway [21, 22].

Table 1.

Metabolomic study in humans in response to environmental pollutants

Pollutant Study population Sample Analytical method Metabolic response Reference
Biphenol A Korean Research project on the Integrated Exposure Assessment of Hazardous Substances for Food Safety (KRIEFS) Urine Non-targeted, UHPLC-Q/TOF MS Significant disturbance in fatty acid elongation and sphingolipid metabolism. Females in 40’s showed elevated inflammatory metabolites: 6-ketoprostaglandin E1 and thromboxane. [86]
Dioxin Czech chemical workers Urine Non-targeted, UHPLC-Q/TOF MS Glucuro- and sulfo-conjugated endogenous steroid metabolites and bile acids were assessed as biomarkers of acute dioxin toxicity. [39]
PAHs Exposed participants were from a polluted rural area less than 2 km downwind of a large coking plant in China Urine Non-targeted, UHPLC-Q/TOF MS ↑Pyroglutamic acid, 3-Methylhistidine, azelaic acid, decenedioic acid, hydroxytetradecanedioic acid, medium-chain acylcarnitines, decenediolyglucuronide
↓Uric acid
[87]
PFAS Overweight and obese Hispanic children (8–14 years) from urban Los Angeles Plasma Non-targeted, UHPLC-Q Exactive MS Significant alterations of lipids (e.g., glycosphingolipids, linoleic acid, and de novo lipogenesis), and amino acids (e.g., aspartate and asparagine, tyrosine, arginine and proline) [42]
Air Pollution Two randomized crossover trials were used including the Oxford Street II (London) and the TAPAS II (Barcelona) studies Serum Non-targeted, UHPLC-Q/TOF MS 17 and 30 metabolic compounds were changed in the Oxford Street II (London) and the TAPAS II (Barcelona) respectively [64]
Ozone Healthy volunteers were exposed to filtered air and ozone Bronchoalveolar lavage fluid Non-targeted, UHPLC-MS Total of 28 and 41 metabolites were differentially expressed after 1-hour and 24-hour post-exposure respectively. [88]
PM 2.5 Healthy volunteers were exposed to PM 2.5 Serum Non-targeted, GC-MS, UHPLC-Q/TOF MS ↑Cortisol, cortisone, epinephrine, norepinephrine, phenylalanine, tyrosine, tetrahydropteridine, L-Tryptophan, N-Acetylserotonin, melatonin
↓Serotonin
[66]
Benzene Painting workers from China Plasma Non-targeted and targeted, HPLC-Q/TOF MS ↑Pyroglutamic acid, glycoursodeoxycholic, linolenyl carnitine, lysoPC(16:1), Bilirubin, LysoPC(18:4/0:0), LysoPC (P-18:0), LysoPC (20:2)
↓L-camitine
[69]
Traffic pollution Participants in the Dorm Room Inhalation to Vehicle Emission (DRIVE) study Plasma, saliva Non-targeted, UHPLC-Q Exactive MS Biological perturbations associated with traffic pollutants included metabolic signaling related to oxidative stress, inflammation, and nucleic acid damage and repair. [68]
Arsenic Health Effects of Arsenic Longitudinal Study (HEALS) Urine Non-targeted, GC-MS 31 molecular features and urinary arsenic was significantly related to 74 molecular features. Six metabolites were significantly associated with either water arsenic or urinary arsenic, including 1,2-dithiane-4,5-diol, L-Threonine, phosphoricacid, pyroglutamic acid, (R*, S*)-3,4-Dihydroxybutanoic acid, and succinic acid. [82]
Cadmium Participants from south China with high Cd exposure Urine Non-targeted, GC-MS 27 known metabolites involved in glucose, amino acid, energy, fatty acid, bone metabolism and TCA cycle were markedly changed [79]
Lead Residents in a village near a used lead-acid battery (ULAB) recycling facility in Japan Urine Targeted, LC-QTRAP MS Ten candidate biomarkers were associated with ATP-binding cassette (ABC) transporters, the disruption of small-molecule transport in the kidney; amino acid, porphyrin, and chlorophyll metabolism; and the heme biosynthetic pathway. [81]

Metabolomics is viewed as a functional readout of other omics techniques. For example, as organisms respond to external stressors, resulting changes to gene expression and protein production occur. These expression-related changes are subjected to a variety of homeostatic controls and feedback mechanisms, which could ultimately be reflected on the endpoint of metabolite formation. This may result in metabolomics being a sensitive indicator of the external stressors compared with other omics technologies. The Human Metabolome Database 4.0 (www.hmdb.ca) lists 114,100 metabolites [23], but the actual number of metabolites could be higher. As analytical techniques and detection sensitivity continue to improve, more metabolites will be identified. Metabolomics has potential as a sensitive technique that is capable of characterizing biological changes within a cell, tissue, or biofluid of an organism in response to environmental pollutant exposure.

In this mini review, we summarize recent metabolomics studies focusing on the association between human diseases and exposure to environmental pollutants, which are grouped based on chemical-physical properties, i.e., persistent organic pollutants, air pollutants, and heavy metals. Identification of novel mechanisms of toxicity using metabolomics are also discussed.

Metabolomics and persistent organic pollutants

Persistent organic pollutants (POPs) are highly lipophilic compounds that are resistant to degradation and tend to bioaccumulate in the environment, food chains and living organisms. Examples of these compounds include dioxins, dichlorodiphenyltrichloroethane (DDT), polychlorinated biphenyls (PCBs), phthalates such as bisphenol A (BPA), and the perfluorinated and polyfluorinated alkyl substances (PFAS). The major route of exposure to POPs is ingestion, which could occur through the consumption of animal derived foods, or through contaminated drinking water In vivo and/or in vitro experiments have shown that even at the low levels reported for non-occupational exposures, these compounds exhibit teratogenicity, immunotoxicity, endocrine toxicity and carcinogenicity [24]. Recent studies show that exposure to POPs have been associated with cardiometabolic risk factors [25, 26]. Epidemiological studies suggest that POPs may act as metabolic disruptors contributing to the growing incidence of metabolic diseases [27, 28].

Metabolomics studies of POPs enables hypothesis generation for hitherto unexplained responses to environmental stressors and thereby helps unravel the underlying mode of action of POPs. Coincident observations across mechanism studies using animal and cell models include impaired gluconeogenesis, altered glucose production and lipid metabolism [2931]. Our laboratory has previously demonstrated that exposure to PCB 126, the most potent dioxin-like PCB, can increase peripheral inflammation in a liver injury mouse model [32]. Metabolites in mice livers were profiled using non-targeted metabolomics method to elucidate possible associations between liver injury and PCB 126 promoted cardiometabolic disease risk. The results demonstrated that PCB 126 altered the liver metabolome and aggravated redox stress, which may predispose to cardiometabolic complications [33]. Other studies employed metabolomic techniques to investigate host-microbiota interactions and found that POPs disrupted gut microbiota and host metabolism [14, 34, 35]. For example, Zhang et al. investigated the effect of 2,3,7,8-tetrachlorodibenzofuran (TCDF) on gut microbiota and host metabolism using NMR and targeted LC-MS [14]. They found that TCDF altered gut microbiota composition and this was associated with altered bile acid metabolism. Additionally, TCDF triggered host metabolic disorders as a result of altered hepatic lipogenesis, gluconeogenesis, and glycogenolysis [14]. Using non-targeted metabolomics, it was found that PCB 126 disrupted the gut microbiota and host metabolism in mice, which imply that the toxic effects of PCB 126 may be initiated in the gut, and the modulation of gut microbiota could be a sensitive marker of PCB 126 exposures [34]. A follow-up study demonstrated that fecal microbe exposed to PCB 126 resulted in reduced succinic acid production, but increased propionate production, both of which can influence host glucose and lipid metabolism [36]. These findings provide new insights into the biochemical consequences of POPs exposure involving the alteration of the gut microbiota and disruption of host metabolism.

Metabolomic-based epidemiologic studies help understand how environmental pollutants disturb metabolism and help to identify biomarkers related to POPs exposure. For example, Carrizo et al. [37] reported a non-targeted metabolomic analysis of human serum samples associated with exposure levels of POPs and found that sphingolipids and glycerophospholipid lipids were significantly different between high and low POPs exposure groups. This study indicated an imbalance in human lipid metabolism upon POPs exposure, and these observed metabolomic fingerprints may have the potential to be classified as biomarkers of POPs exposure. Jeanneret et al. [38, 39] identified human urinary markers of acute dioxin exposure using non-targeted metabolomic approaches, including glucuro- and sulfo-conjugated endogenous steroids and bile acids. Zbucka-Kretowska et al. [40] revealed that increased level of BPA in the maternal plasma was related to higher level of several endocannabinoids using a non-targeted metabolomic analysis. Further studies demonstrated that BPA inhibited fatty acid amide hydrolase activity and consequently led to a rise of plasma endocannabinoids. These studies provided a novel toxicity mechanism of BPA, which may induce adverse pregnancy outcomes by acting on the endocannabinoid system. PFAS exposure and plasma metabolomic studies demonstrated that higher PFAS exposure in adults and children was associated with dysregulated lipid and amino acid metabolism [4143]. Perturbation of lipid and amino acid pathways have been found to be strongly associated with the risk of developing metabolic diseases including type 2 diabetes [44, 45]. Collectively, these studies indicate that PFAS exposure might cause disturbances in lipid and amino acid metabolism, thereby contributing to increased risk of metabolic diseases.

Metabolomics and air pollutants

Air pollution is a complex mixture of gaseous, volatile, semi-volatile and particulate matter, and its exact composition varies widely. The major route of exposure to air pollution is by inhalation. Some of the commonly measured components of the air pollution mixtures include irritant gases, benzene, and particulate matter (PM) [46]. Additionally, various harmful substances are adsorbed or contained in PM2.5, such as heavy metals, polycyclic aromatic hydrocarbons and POPs. Air pollution is the world’s largest environmental threat to health and is responsible each year for an estimated 6.5 million deaths [47, 48]. A growing number of epidemiological studies showed that air pollution accounts for 19% of all cardiovascular deaths, 23% of all ischemic heart disease deaths, and 21% of all stroke deaths [49]. Air pollution has also been linked to cancers [50], neurologic [51], and respiratory diseases [52]. Furthermore, air pollution effects the immune system and is associated with allergic rhinitis, allergic sensitization, and autoimmunity [53, 54]. The most well established mechanism is oxidative stress caused by air pollutants, followed by pulmonary and systemic inflammation [55].

Metabolomics has been increasingly applied to investigate associations between long- and short-term exposure to air pollution and metabolome changes. Non-targeted and targeted metabolomics strategy were adopted to characterize the overall metabolic changes and relevant toxicological pathways in animal serum and urine [5662]. Purine, amino acid, and lipid metabolism are common pathways that are disturbed after air pollutant exposure [5662]. A study focusing on the pulmonary metabolome in rats demonstrated that phospholipid and sphingolipid metabolism were altered after PM2.5 exposure, which may relate to PM2.5-induced pulmonary inflammation and injury [58]. The detrimental effect of air pollution to the human body is closely associated with lipid metabolism, and disturbed plasma lipid profiles have been observed in several cohorts with exposure to various air pollutants [63, 64]. Results from a non-targeted metabolomic study of three cohorts, each a part of the Cooperative Health Research in the Region of Augsburg, Germany, revealed that changes in lysophosphatidylcholines (LPCs) in human plasma were associated with short-term exposure to air pollutants [64]. This result was in agreement with the findings in animal and cell models, in which LPCs were increased in air pollutant exposed rats and alveolar type II cells compared with corresponding control groups [65]. The researchers further determined that the observed increase may have been due to air pollutant-induced activation of phospholipase A2, which catalyzes the hydrolysis of phosphatidylcholines to LPC [65]. Jeong et al. [21] applied MITM non-targeted metabolomics approach to identify metabolites and pathways that could be associated with both air pollutants and compromised health outcomes using human serum samples. It was found that linoleate metabolism and glycerophospholipid metabolism were common MITM pathways linking ultra-fine particles (UFP) exposure to asthma; glycosphingolipid metabolism linking UFP exposure to cardiovascular disease; and metabolites related to carnitine shuttling linking NO2 exposure to cardio-cerebrovascular disorders. The last association was also observed in a recent short-term traffic-related air pollution study [64]. Li et al. [66] reported a randomized, double-blind, crossover trial of metabolomics analysis on particulate air pollution. The results demonstrated that higher PM exposure led to a significant increase in serum levels of stress hormones, suggesting activations of the hypothalamus-pituitary-adrenal and sympathetic-adrenal-medullary axes, which may contribute to adverse cardiovascular and metabolic effects. Other reported metabolic perturbations associated with air pollution exposure are dysregulated inflammatory and oxidative stress related pathways, including leukotriene, vitamin E, and glutathione metabolism [6769]. These altered pathways could contribute to the tissue damage and systemic inflammation observed in response to air pollutant exposure.

Metabolomics and heavy metals

Heavy metals, such as As, cadmium (Cd), mercury (Hg), copper (Cu), lead (Pb), and manganese (Mn), are ubiquitous and are increasingly introduced into the food chain due to industrial and agricultural practices. For the general population, diet and drinking water are the main sources of heavy metal exposure. Heavy metal toxicity results in damage of the lungs, kidneys, liver and other organs [70]. Additionally, heavy metals can enter the brain through the disruption of blood-brain barrier, subsequently damaging the nervous system [71]. For example, Cd-dependent neurotoxicity has been reported to be related to Alzheimer’s and Parkinson’s diseases, however, the molecular mechanisms of Cd toxicity remain to be clarified [72]. The neurotoxicity of Cd has been investigated in neuronal PC-12 cells by a non-targeted metabolomic approach [73]. The results demonstrated that Cd exposure modulated energy metabolism (glycolysis, TCA cycle and fatty acid β-oxidation), cell membrane composition, as well as the anti-oxidant defense system. Using metabolomic analysis of urine samples, it was demonstrated that Cd disrupted energy and lipid metabolism, as well as induced oxidative stress, which contributed to tissue damage [74, 75]. Chronic exposure of inorganic As has been associated with increased risk for numerous metabolic syndromes [76]. After ingestion, inorganic As is metabolized through methylation and reduction reactions which ultimately produce methylated metabolites, such as monomethylarsonic acid (MMA) and dimethylarsinic acid (DMA). Inter-individual differences in methylation capacity have been associated with risk of health outcomes [77]. Spratlen et al. [78] explored the mechanism that may explain the associations between As metabolism and metabolic outcomes using targeted metabolomic analysis of human plasma. It was found that one carbon metabolism pathway was significantly related with both As metabolism and diabetes-related outcomes. Additionally, association of lower MMA% and higher DMA% with diabetes-related outcomes may be influenced by one carbon metabolism status. Metabolomics has been applied to the identification of biomarkers for heavy metal exposure using human urine samples [7982] (Table 1). For example, Wu et al.[82] investigated associations between reproducible urinary metabolites and As exposure in Bangladesh Adults using non-targeted metabolomics. Six metabolites (1,2-dithiane-4,5-diol, l-threonine, phosphoric acid, pyroglutamic acid, (R*,S*)-3,4-dihydroxybutanoic acid, and succinic acid) were identified as significantly associated with either water concentrations of As or urinary As after adjustment for multiple comparisons. Xu et al. [79] identified urine metabolites in women exposed to high Cd levels using targeted metabolomics method. The results demonstrated that the abundance of 48 metabolites were markedly changed in subjects with high Cd levels exposure, among which 8 endogenous metabolites were identified as potential early candidate biomarkers. Overall, these results provided new insights into the mechanism of heavy metal toxicity and identification of biomarkers associated with exposure.

Conclusions and future perspectives

In this mini-review, we have highlighted the increasing potential of metabolomics for assessing disease risks of environmental pollutants. These studies have enhanced our understanding on how environmental pollutant exposure can alter biological mechanisms and shed light on disease etiology. However, there are still many challenges. For example, human exposure data (including pollutants, lifestyle factors and behaviors) are often limited or incomplete, and the estimation of associated disease risks can be severely hindered. The exposome, which measures all the exposures of an individual over a timeframe, such as a lifetime, has recently been proposed as a new paradigm to encompass the totality of human environmental [83]. Additionally, some metabolites do respond sensitively to environmental stressors, such as metabolites within glycolysis and the TCA cycle, however, many of these metabolites are not specific enough to be used for diagnostic purposes. With the advance of multi-omics technologies, a more comprehensive understanding of disease risks and consequently promising biomarkers that are unique to specific pollutant exposure and diseases are becoming possible. Advancements in next generation sequencing in recent years has allowed detailed genomics, transcriptomics, and bioinformatic analyses of altered biological pathways related to pollutant exposure [84, 85]. These technologies, combined with metabolomics, will provide unprecedented insights into key pathways that connect pollutant exposure to disease risks and pathological endpoints.

Acknowledgements

This work was supported in part by NIEHS/NIH P42ES007380 and K99ES028734. We thank Tom Dolan (Medical Illustration, College of Medicine, University of Kentucky) for preparing Figure 1.

Footnotes

Financial interests

The authors declare they have no actual or potential competing conflict of financial interest relevant to this work.

Reference

  • 1.Ab Manan N, Noor Aizuddin A, Hod R. Effect of Air Pollution and Hospital Admission: A Systematic Review. Ann Glob Health 2018; 84(4): 670–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Khera AV, Emdin CA, Drake I, Natarajan P, Bick AG, Cook NR, Chasman DI, Baber U, Mehran R, Rader DJ, Fuster V, Boerwinkle E, Melander O, Orho-Melander M, Ridker PM, Kathiresan S. Genetic Risk, Adherence to a Healthy Lifestyle, and Coronary Disease. N Engl J Med 2016; 375(24): 2349–2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hennig B, Petriello MC, Gamble MV, Surh YJ, Kresty LA, Frank N, Rangkadilok N, Ruchirawat M, Suk WA. The role of nutrition in influencing mechanisms involved in environmentally mediated diseases. Rev Environ Health 2018; 33(1): 87–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Au WW. Life style factors and acquired susceptibility to environmental disease. Int J Hyg Environ Health 2001; 204(1): 17–22. [DOI] [PubMed] [Google Scholar]
  • 5.Zeliger HI. Co-morditities of environmental diseases: A common cause. Interdiscip Toxicol 2014; 7(3): 117–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Landrigan PJ, Fuller R, Horton R. Environmental pollution, health, and development: a Lancet-Global Alliance on Health and Pollution-Icahn School of Medicine at Mount Sinai Commission. Lancet 2015; 386(10002): 1429–31. [DOI] [PubMed] [Google Scholar]
  • 7.Prüss-Ustün A, Wolf J, Corvalan C, Neville T, Bos R, Neira M. Diseases due to unhealthy environments: an updated estimate of the global burden of disease attributable to environmental determinants of health. J Public Health (Oxf) 2017; 39(3): 464–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Prüss-Ustün A, Wolf J, Corvalán C, Bos R, Neira M. Preventing disease through healthy environments: a global assessment of the burden of disease from environmental risks. World Health Organization 2016. [Google Scholar]
  • 9.Yan M, Xu G. Current and future perspectives of functional metabolomics in disease studies-A review. Anal Chim Acta 2018; 1037: 41–54. [DOI] [PubMed] [Google Scholar]
  • 10.Amberg A, Riefke B, Schlotterbeck G, Ross A, Senn H, Dieterle F, Keck M. NMR and MS Methods for Metabolomics. Methods Mol Biol 2017; 1641: 229–258. [DOI] [PubMed] [Google Scholar]
  • 11.Kumar B, Prakash A, Ruhela RK, Medhi B. Potential of metabolomics in preclinical and clinical drug development. Pharmacol Rep 2014; 66(6): 956–63. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang L, Hatzakis E, Nichols RG, Hao R, Correll J, Smith PB, Chiaro CR, Perdew GH, Patterson AD. Metabolomics Reveals that Aryl Hydrocarbon Receptor Activation by Environmental Chemicals Induces Systemic Metabolic Dysfunction in Mice. Environ Sci Technol 2015; 49(13): 8067–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Huang MC, Douillet C, Su M, Zhou K, Wu T, Chen W, Galanko JA, Drobna Z, Saunders RJ, Martin E, Fry RC, Jia W, Styblo M. Metabolomic profiles of arsenic (+3 oxidation state) methyltransferase knockout mice: effect of sex and arsenic exposure. Arch Toxicol 2017; 91(1): 189–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhang L, Nichols RG, Correll J, Murray IA, Tanaka N, Smith PB, Hubbard TD, Sebastian A, Albert I, Hatzakis E, Gonzalez FJ, Perdew GH, Patterson AD. Persistent Organic Pollutants Modify Gut Microbiota-Host Metabolic Homeostasis in Mice Through Aryl Hydrocarbon Receptor Activation. Environ Health Perspect 2015; 123(7): 679–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Xue J, Lai Y, Chi L, Tu P, Leng J, Liu CW, Ru H, Lu K. Serum Metabolomics Reveals That Gut Microbiome Perturbation Mediates Metabolic Disruption Induced by Arsenic Exposure in Mice. J Proteome Res 2019; 18(3): 1006–1018. [DOI] [PubMed] [Google Scholar]
  • 16.Kim SJ, Heo SH, Lee DS, Hwang IG, Lee YB, Cho HY. Gender differences in pharmacokinetics and tissue distribution of 3 perfluoroalkyl and polyfluoroalkyl substances in rats. Food Chem Toxicol 2016; 97: 243–255. [DOI] [PubMed] [Google Scholar]
  • 17.Kundakovic M, Gudsnuk K, Herbstman JB, Tang D, Perera FP, Champagne FA. DNA methylation of BDNF as a biomarker of early-life adversity. Proc Natl Acad Sci U S A 2015; 112(22): 6807–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Leasure JL, Giddabasappa A, Chaney S, Johnson JE Jr., Pothakos K, Lau YS, Fox DA. Low-level human equivalent gestational lead exposure produces sex-specific motor and coordination abnormalities and late-onset obesity in year-old mice. Environ Health Perspect 2008; 116(3): 355–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fearnley LG, Inouye M. Metabolomics in epidemiology: from metabolite concentrations to integrative reaction networks. Int J Epidemiol 2016; 45(5): 1319–1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vineis P, van Veldhoven K, Chadeau-Hyam M, Athersuch TJ. Advancing the application of omics-based biomarkers in environmental epidemiology. Environ Mol Mutagen 2013; 54(7): 461–7. [DOI] [PubMed] [Google Scholar]
  • 21.Jeong A, Fiorito G, Keski-Rahkonen P, Imboden M, Kiss A, Robinot N, Gmuender H, Vlaanderen J, Vermeulen R, Kyrtopoulos S, Herceg Z, Ghantous A, Lovison G, Galassi C, Ranzi A, Krogh V, Grioni S, Agnoli C, Sacerdote C, Mostafavi N, Naccarati A, Scalbert A, Vineis P, Probst-Hensch N, Consortium EX. Perturbation of metabolic pathways mediates the association of air pollutants with asthma and cardiovascular diseases. Environ Int 2018; 119: 334–345. [DOI] [PubMed] [Google Scholar]
  • 22.Chadeau-Hyam M, Athersuch TJ, Keun HC, De Iorio M, Ebbels TM, Jenab M, Sacerdote C, Bruce SJ, Holmes E, Vineis P. Meeting-in-the-middle using metabolic profiling - a strategy for the identification of intermediate biomarkers in cohort studies. Biomarkers 2011; 16(1): 83–8. [DOI] [PubMed] [Google Scholar]
  • 23.Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vazquez-Fresno R, Sajed T, Johnson D, Li C, Karu N, Sayeeda Z, Lo E, Assempour N, Berjanskii M, Singhal S, Arndt D, Liang Y, Badran H, Grant J, Serra-Cayuela A, Liu Y, Mandal R, Neveu V, Pon A, Knox C, Wilson M, Manach C, Scalbert A. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res 2018; 46(D1): D608–D617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ashraf MA. Persistent organic pollutants (POPs): a global issue, a global challenge. Environ Sci Pollut Res Int 2017; 24(5): 4223–4227. [DOI] [PubMed] [Google Scholar]
  • 25.Wu H, Bertrand KA, Choi AL, Hu FB, Laden F, Grandjean P, Sun Q. Persistent organic pollutants and type 2 diabetes: a prospective analysis in the nurses’ health study and meta-analysis. Environ Health Perspect 2013; 121(2): 153–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dusanov S, Ruzzin J, Kiviranta H, Klemsdal TO, Retterstol L, Rantakokko P, Airaksinen R, Djurovic S, Tonstad S. Associations between persistent organic pollutants and metabolic syndrome in morbidly obese individuals. Nutr Metab Cardiovasc Dis 2018; 28(7): 735–742. [DOI] [PubMed] [Google Scholar]
  • 27.Heindel JJ, Blumberg B, Cave M, Machtinger R, Mantovani A, Mendez MA, Nadal A, Palanza P, Panzica G, Sargis R, Vandenberg LN, Vom Saal F. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol 2017; 68: 3–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dusanov S, Ruzzin J, Kiviranta H, Klemsdal TO, Retterstøl L, Rantakokko P, Airaksinen R, Djurovic S, Tonstad S. Associations between persistent organic pollutants and metabolic syndrome in morbidly obese individuals. Nutrition, Metabolism and Cardiovascular Diseases 2018; 28(7): 735–742. [DOI] [PubMed] [Google Scholar]
  • 29.Gadupudi GS, Klaren WD, Olivier AK, Klingelhutz AJ, Robertson LW. PCB126-Induced Disruption in Gluconeogenesis and Fatty Acid Oxidation Precedes Fatty Liver in Male Rats. Toxicol Sci 2016; 149(1): 98–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wu X, Yang J, Morisseau C, Robertson LW, Hammock B, Lehmler HJ. 3,3’,4,4’,5-Pentachlorobiphenyl (PCB 126) Decreases Hepatic and Systemic Ratios of Epoxide to Diol Metabolites of Unsaturated Fatty Acids in Male Rats. Toxicol Sci 2016; 152(2): 309–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wu H, Yu W, Meng F, Mi J, Peng J, Liu J, Zhang X, Hai C, Wang X. Polychlorinated biphenyls-153 induces metabolic dysfunction through activation of ROS/NF-kappaB signaling via downregulation of HNF1b. Redox Biol 2017; 12: 300–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wahlang B, Barney J, Thompson B, Wang C, Hamad OM, Hoffman JB, Petriello MC, Morris AJ, Hennig B. Editor’s Highlight: PCB126 Exposure Increases Risk for Peripheral Vascular Diseases in a Liver Injury Mouse Model. Toxicol Sci 2017; 160(2): 256–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Deng P, Barney J, Petriello MC, Morris AJ, Wahlang B, Hennig B. Hepatic metabolomics reveals that liver injury increases PCB 126-induced oxidative stress and metabolic dysfunction. Chemosphere 2019; 217: 140–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Petriello MC, Hoffman JB, Vsevolozhskaya O, Morris AJ, Hennig B. Dioxin-like PCB 126 increases intestinal inflammation and disrupts gut microbiota and metabolic homeostasis. Environ Pollut 2018; 242(Pt A): 1022–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhang L, Nichols RG, Patterson AD. The aryl hydrocarbon receptor as a moderator of host-microbiota communication. Curr Opin Toxicol 2017; 2: 30–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hoffman JB, Flythe MD, Hennig B. Environmental pollutant-mediated disruption of gut microbial metabolism of the prebiotic inulin. Anaerobe 2019; 55: 96–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Carrizo D, Chevallier OP, Woodside JV, Brennan SF, Cantwell MM, Cuskelly G, Elliott CT. Untargeted metabolomic analysis of human serum samples associated with exposure levels of Persistent organic pollutants indicate important perturbations in Sphingolipids and Glycerophospholipids levels. Chemosphere 2017; 168: 731–738. [DOI] [PubMed] [Google Scholar]
  • 38.Jeanneret F, Tonoli D, Hochstrasser D, Saurat JH, Sorg O, Boccard J, Rudaz S. Evaluation and identification of dioxin exposure biomarkers in human urine by high-resolution metabolomics, multivariate analysis and in vitro synthesis. Toxicol Lett 2016; 240(1): 22–31. [DOI] [PubMed] [Google Scholar]
  • 39.Jeanneret F, Boccard J, Badoud F, Sorg O, Tonoli D, Pelclova D, Vlckova S, Rutledge DN, Samer CF, Hochstrasser D, Saurat JH, Rudaz S. Human urinary biomarkers of dioxin exposure: analysis by metabolomics and biologically driven data dimensionality reduction. Toxicol Lett 2014; 230(2): 234–43. [DOI] [PubMed] [Google Scholar]
  • 40.Zbucka-Kretowska M, Zbucki R, Parfieniuk E, Maslyk M, Lazarek U, Miltyk W, Czerniecki J, Wolczynski S, Kretowski A, Ciborowski M. Evaluation of Bisphenol A influence on endocannabinoid system in pregnant women. Chemosphere 2018; 203: 387–392. [DOI] [PubMed] [Google Scholar]
  • 41.Salihovic S, Fall T, Ganna A, Broeckling CD, Prenni JE, Hyotylainen T, Karrman A, Lind PM, Ingelsson E, Lind L. Identification of metabolic profiles associated with human exposure to perfluoroalkyl substances. J Expo Sci Environ Epidemiol 2019; 29(2): 196–205. [DOI] [PubMed] [Google Scholar]
  • 42.Alderete TL, Jin R, Walker DI, Valvi D, Chen Z, Jones DP, Peng C, Gilliland FD, Berhane K, Conti DV, Goran MI, Chatzi L. Perfluoroalkyl substances, metabolomic profiling, and alterations in glucose homeostasis among overweight and obese Hispanic children: A proof-of-concept analysis. Environ Int 2019; 126: 445–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kingsley SL, Walker DI, Calafat AM, Chen A, Papandonatos GD, Xu Y, Jones DP, Lanphear BP, Pennell KD, Braun JM. Metabolomics of childhood exposure to perfluoroalkyl substances: a cross-sectional study. Metabolomics 2019; 15(7): 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Guasch-Ferre M, Hruby A, Toledo E, Clish CB, Martinez-Gonzalez MA, Salas-Salvado J, Hu FB. Metabolomics in Prediabetes and Diabetes: A Systematic Review and Meta-analysis. Diabetes Care 2016; 39(5): 833–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Newgard CB. Metabolomics and Metabolic Diseases: Where Do We Stand? Cell Metab 2017; 25(1): 43–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Langrish JP, Bosson J, Unosson J, Muala A, Newby DE, Mills NL, Blomberg A, Sandstrom T. Cardiovascular effects of particulate air pollution exposure: time course and underlying mechanisms. J Intern Med 2012; 272(3): 224–39. [DOI] [PubMed] [Google Scholar]
  • 47.Campbell-Lendrum D, Pruss-Ustun A. Climate change, air pollution and noncommunicable diseases. Bull World Health Organ 2019; 97(2): 160–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Landrigan PJ, Fuller R, Acosta NJR, Adeyi O, Arnold R, Basu NN, Balde AB, Bertollini R, Bose-O’Reilly S, Boufford JI, Breysse PN, Chiles T, Mahidol C, Coll-Seck AM, Cropper ML, Fobil J, Fuster V, Greenstone M, Haines A, Hanrahan D, Hunter D, Khare M, Krupnick A, Lanphear B, Lohani B, Martin K, Mathiasen KV, McTeer MA, Murray CJL, Ndahimananjara JD, Perera F, Potocnik J, Preker AS, Ramesh J, Rockstrom J, Salinas C, Samson LD, Sandilya K, Sly PD, Smith KR, Steiner A, Stewart RB, Suk WA, van Schayck OCP, Yadama GN, Yumkella K, Zhong M. The Lancet Commission on pollution and health. Lancet 2018; 391(10119): 462–512. [DOI] [PubMed] [Google Scholar]
  • 49.Hadley MB, Baumgartner J, Vedanthan R. Developing a Clinical Approach to Air Pollution and Cardiovascular Health. Circulation 2018; 137(7): 725–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lavigne E, Belair MA, Do MT, Stieb DM, Hystad P, van Donkelaar A, Martin RV, Crouse DL, Crighton E, Chen H, Brook JR, Burnett RT, Weichenthal S, Villeneuve PJ, To T, Cakmak S, Johnson M, Yasseen AS 3rd, Johnson KC, Ofner M, Xie L, Walker M. Maternal exposure to ambient air pollution and risk of early childhood cancers: A population-based study in Ontario, Canada. Environ Int 2017; 100: 139–147. [DOI] [PubMed] [Google Scholar]
  • 51.Chen H, Kwong JC, Copes R, Tu K, Villeneuve PJ, van Donkelaar A, Hystad P, Martin RV, Murray BJ, Jessiman B, Wilton AS, Kopp A, Burnett RT. Living near major roads and the incidence of dementia, Parkinson’s disease, and multiple sclerosis: a population-based cohort study. Lancet 2017; 389(10070): 718–726. [DOI] [PubMed] [Google Scholar]
  • 52.Schraufnagel DE, Balmes JR, Cowl CT, De Matteis S, Jung SH, Mortimer K, Perez-Padilla R, Rice MB, Riojas-Rodriguez H, Sood A, Thurston GD, To T, Vanker A, Wuebbles DJ. Air Pollution and Noncommunicable Diseases: A Review by the Forum of International Respiratory Societies’ Environmental Committee, Part 2: Air Pollution and Organ Systems. Chest 2019; 155(2): 417–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bernatsky S, Smargiassi A, Barnabe C, Svenson LW, Brand A, Martin RV, Hudson M, Clarke AE, Fortin PR, van Donkelaar A, Edworthy S, Belisle P, Joseph L. Fine particulate air pollution and systemic autoimmune rheumatic disease in two Canadian provinces. Environ Res 2016; 146: 85–91. [DOI] [PubMed] [Google Scholar]
  • 54.Bernatsky S, Smargiassi A, Johnson M, Kaplan GG, Barnabe C, Svenson L, Brand A, Bertazzon S, Hudson M, Clarke AE, Fortin PR, Edworthy S, Belisle P, Joseph L. Fine particulate air pollution, nitrogen dioxide, and systemic autoimmune rheumatic disease in Calgary, Alberta. Environ Res 2015; 140: 474–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lodovici M, Bigagli E. Oxidative stress and air pollution exposure. J Toxicol 2011; 2011: 487074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang Y, Li Y, Shi Z, Wu J, Yang X, Feng L, Ren L, Duan J, Sun Z. Metabolic impact induced by total, water soluble and insoluble components of PM2.5 acute exposure in mice. Chemosphere 2018; 207: 337–346. [DOI] [PubMed] [Google Scholar]
  • 57.Zhang Y, Hu H, Shi Y, Yang X, Cao L, Wu J, Asweto CO, Feng L, Duan J, Sun Z. (1)H NMR-based metabolomics study on repeat dose toxicity of fine particulate matter in rats after intratracheal instillation. Sci Total Environ 2017; 589: 212–221. [DOI] [PubMed] [Google Scholar]
  • 58.Wang X, Jiang S, Liu Y, Du X, Zhang W, Zhang J, Shen H. Comprehensive pulmonary metabolome responses to intratracheal instillation of airborne fine particulate matter in rats. Sci Total Environ 2017; 592: 41–50. [DOI] [PubMed] [Google Scholar]
  • 59.Sun R, Zhang J, Xiong M, Chen Y, Yin L, Pu Y. Metabonomics biomarkers for subacute toxicity screening for benzene exposure in mice. J Toxicol Environ Health A 2012; 75(18): 1163–73. [DOI] [PubMed] [Google Scholar]
  • 60.Brower JB, Doyle-Eisele M, Moeller B, Stirdivant S, McDonald JD, Campen MJ. Metabolomic changes in murine serum following inhalation exposure to gasoline and diesel engine emissions. Inhal Toxicol 2016; 28(5): 241–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhao C, Niu M, Song S, Li J, Su Z, Wang Y, Gao Q, Wang H. Serum metabolomics analysis of mice that received repeated airway exposure to a water-soluble PM2.5 extract. Ecotoxicol Environ Saf 2019; 168: 102–109. [DOI] [PubMed] [Google Scholar]
  • 62.Miller DB, Karoly ED, Jones JC, Ward WO, Vallanat BD, Andrews DL, Schladweiler MC, Snow SJ, Bass VL, Richards JE, Ghio AJ, Cascio WE, Ledbetter AD, Kodavanti UP. Inhaled ozone (O3)-induces changes in serum metabolomic and liver transcriptomic profiles in rats. Toxicol Appl Pharmacol 2015; 286(2): 65–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Breitner S, Schneider A, Devlin RB, Ward-Caviness CK, Diaz-Sanchez D, Neas LM, Cascio WE, Peters A, Hauser ER, Shah SH, Kraus WE. Associations among plasma metabolite levels and short-term exposure to PM2.5 and ozone in a cardiac catheterization cohort. Environ Int 2016; 97: 76–84. [DOI] [PubMed] [Google Scholar]
  • 64.van Veldhoven K, Kiss A, Keski-Rahkonen P, Robinot N, Scalbert A, Cullinan P, Chung KF, Collins P, Sinharay R, Barratt BM, Nieuwenhuijsen M, Rodoreda AA, Carrasco-Turigas G, Vlaanderen J, Vermeulen R, Portengen L, Kyrtopoulos SA, Ponzi E, Chadeau-Hyam M, Vineis P. Impact of short-term traffic-related air pollution on the metabolome - Results from two metabolome-wide experimental studies. Environ Int 2019; 123: 124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhang SY, Shao D, Liu H, Feng J, Feng B, Song X, Zhao Q, Chu M, Jiang C, Huang W, Wang X. Metabolomics analysis reveals that benzo[a]pyrene, a component of PM2.5, promotes pulmonary injury by modifying lipid metabolism in a phospholipase A2-dependent manner in vivo and in vitro. Redox Biol 2017; 13: 459–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Li H, Cai J, Chen R, Zhao Z, Ying Z, Wang L, Chen J, Hao K, Kinney PL, Chen H, Kan H. Particulate Matter Exposure and Stress Hormone Levels: A Randomized, Double-Blind, Crossover Trial of Air Purification. Circulation 2017; 136(7): 618–627. [DOI] [PubMed] [Google Scholar]
  • 67.Ladva CN, Golan R, Liang D, Greenwald R, Walker DI, Uppal K, Raysoni AU, Tran V, Yu T, Flanders WD, Miller GW, Jones DP, Sarnat JA. Particulate metal exposures induce plasma metabolome changes in a commuter panel study. PLoS One 2018; 13(9): e0203468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liang D, Moutinho JL, Golan R, Yu T, Ladva CN, Niedzwiecki M, Walker DI, Sarnat SE, Chang HH, Greenwald R, Jones DP, Russell AG, Sarnat JA. Use of high-resolution metabolomics for the identification of metabolic signals associated with traffic-related air pollution. Environ Int 2018; 120: 145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sun R, Xu K, Zhang Q, Jiang X, Man Z, Yin L, Zhang J, Pu Y. Plasma metabonomics investigation reveals involvement of fatty acid oxidation in hematotoxicity in Chinese benzene-exposed workers with low white blood cell count. Environ Sci Pollut Res Int 2018; 25(32): 32506–32514. [DOI] [PubMed] [Google Scholar]
  • 70.Jaishankar M, Tseten T, Anbalagan N, Mathew BB, Beeregowda KN. Toxicity, mechanism and health effects of some heavy metals. Interdiscip Toxicol 2014; 7(2): 60–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Caito S, Aschner M. Neurotoxicity of metals. Handb Clin Neurol 2015; 131: 169–89. [DOI] [PubMed] [Google Scholar]
  • 72.Branca JJV, Morucci G, Pacini A. Cadmium-induced neurotoxicity: still much ado. Neural Regen Res 2018; 13(11): 1879–1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zong L, Xing J, Liu S, Liu Z, Song F. Cell metabolomics reveals the neurotoxicity mechanism of cadmium in PC12 cells. Ecotoxicol Environ Saf 2018; 147: 26–33. [DOI] [PubMed] [Google Scholar]
  • 74.Sarma SN, Saleem A, Lee JY, Tokumoto M, Hwang GW, Man Chan H, Satoh M. Effects of long-term cadmium exposure on urinary metabolite profiles in mice. J Toxicol Sci 2018; 43(2): 89–100. [DOI] [PubMed] [Google Scholar]
  • 75.Chen S, Zhang M, Bo L, Li S, Hu L, Zhao X, Sun C. Metabolomic analysis of the toxic effect of chronic exposure of cadmium on rat urine. Environ Sci Pollut Res Int 2018; 25(4): 3765–3774. [DOI] [PubMed] [Google Scholar]
  • 76.Moon K, Guallar E, Navas-Acien A. Arsenic exposure and cardiovascular disease: an updated systematic review. Curr Atheroscler Rep 2012; 14(6): 542–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chen Y, Wu F, Liu M, Parvez F, Slavkovich V, Eunus M, Ahmed A, Argos M, Islam T, Rakibuz-Zaman M, Hasan R, Sarwar G, Levy D, Graziano J, Ahsan H. A prospective study of arsenic exposure, arsenic methylation capacity, and risk of cardiovascular disease in Bangladesh. Environ Health Perspect 2013; 121(7): 832–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Spratlen MJ, Grau-Perez M, Umans JG, Yracheta J, Best LG, Francesconi K, Goessler W, Bottiglieri T, Gamble MV, Cole SA, Zhao J, Navas-Acien A. Targeted metabolomics to understand the association between arsenic metabolism and diabetes-related outcomes: Preliminary evidence from the Strong Heart Family Study. Environ Res 2019; 168: 146–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Xu Y, Wang J, Liang X, Gao Y, Chen W, Huang Q, Liang C, Tang L, Ouyang G, Yang X. Urine metabolomics of women from small villages exposed to high environmental cadmium levels. Environ Toxicol Chem 2016; 35(5): 1268–75. [DOI] [PubMed] [Google Scholar]
  • 80.Baker MG, Simpson CD, Lin YS, Shireman LM, Seixas N. The Use of Metabolomics to Identify Biological Signatures of Manganese Exposure. Ann Work Expo Health 2017; 61(4): 406–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Eguchi A, Nomiyama K, Sakurai K, Kim Trang PT, Viet PH, Takahashi S, Iwata H, Tanabe S, Todaka E, Mori C. Alterations in urinary metabolomic profiles due to lead exposure from a lead-acid battery recycling site. Environ Pollut 2018; 242(Pt A): 98–105. [DOI] [PubMed] [Google Scholar]
  • 82.Wu F, Chi L, Ru H, Parvez F, Slavkovich V, Eunus M, Ahmed A, Islam T, Rakibuz-Zaman M, Hasan R, Sarwar G, Graziano JH, Ahsan H, Lu K, Chen Y. Arsenic Exposure from Drinking Water and Urinary Metabolomics: Associations and Long-Term Reproducibility in Bangladesh Adults. Environ Health Perspect 2018; 126(1): 017005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Beans C. News Feature: Exposing the exposome to elucidate disease. Proc Natl Acad Sci U S A 2018; 115(47): 11859–11862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fave MJ, Lamaze FC, Soave D, Hodgkinson A, Gauvin H, Bruat V, Grenier JC, Gbeha E, Skead K, Smargiassi A, Johnson M, Idaghdour Y, Awadalla P. Gene-by-environment interactions in urban populations modulate risk phenotypes. Nat Commun 2018; 9(1): 827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Karoui A, Crochemore C, Mulder P, Preterre D, Cazier F, Dewaele D, Corbiere C, Mekki M, Vendeville C, Richard V, Vaugeois JM, Fardel O, Sichel F, Lecureur V, Monteil C. An integrated functional and transcriptomic analysis reveals that repeated exposure to diesel exhaust induces sustained mitochondrial and cardiac dysfunctions. Environ Pollut 2019; 246: 518–526. [DOI] [PubMed] [Google Scholar]
  • 86.Cho S, Khan A, Jee SH, Lee HS, Hwang MS, Koo YE, Park YH. High resolution metabolomics to determines the risk associated with bisphenol A exposure in humans. Environ Toxicol Pharmacol 2018; 58: 1–10. [DOI] [PubMed] [Google Scholar]
  • 87.Wang Z, Zheng Y, Zhao B, Zhang Y, Liu Z, Xu J, Chen Y, Yang Z, Wang F, Wang H, He J, Zhang R, Abliz Z. Human metabolic responses to chronic environmental polycyclic aromatic hydrocarbon exposure by a metabolomic approach. J Proteome Res 2015; 14(6): 2583–93. [DOI] [PubMed] [Google Scholar]
  • 88.Cheng W, Duncan KE, Ghio AJ, Ward-Caviness C, Karoly ED, Diaz-Sanchez D, Conolly RB, Devlin RB. Changes in Metabolites Present in Lung-Lining Fluid Following Exposure of Humans to Ozone. Toxicol Sci 2018; 163(2): 430–439. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES