Next Article in Journal
Pharmacological Therapy of Non-Alcoholic Fatty Liver Disease: What Drugs Are Available Now and Future Perspectives
Next Article in Special Issue
Determination of Triglycidyl Isocyanurate in Workplace Air
Previous Article in Journal
Risk Communication and Factors Influencing Private Well Testing Behavior: A Systematic Scoping Review
Previous Article in Special Issue
Assessment of Occupational Safety and Hygiene Perception among Afro-Caribbean Hair Salon Operators in Manchester, United Kingdom
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genomics of Particulate Matter Exposure Associated Cardiopulmonary Disease: A Narrative Review

1
Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, NYU, School of Medicine, New York, NY 10016, USA
2
Department of Environmental Medicine, New York University, School of Medicine, New York, NY 10016, USA
3
Bureau of Health Services, Fire Department of New York, Brooklyn, NY 11201, USA
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2019, 16(22), 4335; https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph16224335
Submission received: 23 September 2019 / Revised: 28 October 2019 / Accepted: 30 October 2019 / Published: 7 November 2019
(This article belongs to the Special Issue Air Pollution: Occupational Exposure and Public Health)

Abstract

:
Particulate matter (PM) exposure is associated with the development of cardiopulmonary disease. Our group has studied the adverse health effects of World Trade Center particulate matter (WTC-PM) exposure on firefighters. To fully understand the complex interplay between exposure, organism, and resultant disease phenotype, it is vital to analyze the underlying role of genomics in mediating this relationship. A PubMed search was performed focused on environmental exposure, genomics, and cardiopulmonary disease. We included original research published within 10 years, on epigenetic modifications and specific genetic or allelic variants. The initial search resulted in 95 studies. We excluded manuscripts that focused on work-related chemicals, heavy metals and tobacco smoke as primary sources of exposure, as well as reviews, prenatal research, and secondary research studies. Seven full-text articles met pre-determined inclusion criteria, and were reviewed. The effects of air pollution were evaluated in terms of methylation (n = 3), oxidative stress (n = 2), and genetic variants (n = 2). There is evidence to suggest that genomics plays a meditating role in the formation of adverse cardiopulmonary symptoms and diseases that surface after exposure events. Genomic modifications and variations affect the association between environmental exposure and cardiopulmonary disease, but additional research is needed to further define this relationship.

1. Introduction

The adverse effects of particulate matter (PM) exposure are a global health concern. PM is a heterogeneous mixture classified by aerodynamic diameter as fine (≤2.5 µm) or coarse (2.5–10 µm) [1]. Respirable PM can cause damage to multiple organs, primarily to the lungs [2,3]. Wood smoke, black carbon, and traffic-related air pollutants (TRAP) have also been associated with increased risk of lung injury [4,5,6,7]. Concern of exposure to PM and other air pollutants was heightened after the events that took place on September 11th, 2001 (9/11). More than 300,000 workers, local residents, and rescue workers were exposed to about 10 million tons of material that was aerosolized after the collapse of the World Trade Center (WTC) [8]. World Trade Center particulate matter (WTC-PM) differed from ambient PM in composition and in its significantly higher concentration; it was comprised of debris from construction buildings containing concrete, pulverized glass, alkaline metals, asbestos, and components of jet fuel [9,10].
Our group has extensively examined the effects of WTC-PM on firefighters. Although we have shown multiple mediators linking exposure and WTC lung injury (WTC-LI; defined as FEV1% predicted less than the lower limit of normal), there is a lack of knowledge of how genomics mediates this relationship [8,11,12,13]. Genomics research analyzes the genome, aiming to understand its function and mark changes [14]. This work has provided vital information regarding disease development and continues remains important in the early detection of disease [15,16,17,18]. The association between genetic variations, epigenetic markers, and risk of disease is a developing area of research. Since the genome can be affected by environmental interactions, knowledge of the relationship between the genome and exposure to air pollutants can help to establish future preventative and therapeutic measures [19].
Recent studies have dissected the relationship between the genome and disease state after adverse environmental exposure events [20,21,22]. Within a cohort of subjects present during the WTC attacks, varying expression of genes involved in the hypothalamic-pituitary-adrenal axis was linked to post-traumatic stress disorder [23]. A study of firefighters exposed to smoke and debris identified 17 variants of human leukocyte antigen (HLA) and non-HLA genes associated with sarcoidosis [21]. These studies highlight the importance of genomics as a key component of the systems biology approach of disease understanding. However, there is limited research on the specific association between exposure to air pollution and genomic effects as it relates to cardiopulmonary disease. Cardiopulmonary disease is of concern, due to the rising number of cases in PM-exposed populations [9,24,25,26]. The purpose of this manuscript is to review the literature published over the prior 10 years that investigate the genomics of PM-exposure related cardiopulmonary disease. This review will help advance subsequent studies examining WTC-PM exposure, which includes PM of a heterogeneous aerodynamic diameter. This is in contrast to other more recent reviews on general pollution (including those in soil and water), or fine particle environmental pollution, and cardiovascular disease [27,28].

2. Methods

2.1. Eligibility/Study Selection

PubMed was searched on 6 November 2019, for the Search Terms as per Table 1. Results of this Search (n = 95) are in Supplemental Table S1. Titles and abstracts (n = 65) screened by AN, JC, and EW, Supplemental Table S2.

2.2. Definitions

Genomic modification was defined as any genetic or allelic variants, as well as signs of oxidative stress marked at specific loci. Epigenetic modification was defined as the addition or deletion of epigenetic markers that altered the expression of cardiac or respiratory related genes. In this review, cardiopulmonary disease was defined to include obstructive airways disease (OAD), chronic obstructive pulmonary disease (COPD), asthma, chronic bronchitis, emphysema, heart failure, coronary artery disease, congenital heart disease, and peripheral artery disease. Environmental exposure was defined as non-tobacco smoke, PM, and TRAP.

2.3. Search Terms

PubMed was queried (06/11/2019) for publications about genomics in relation to cardiopulmonary disease and exposure to air pollutants, utilizing the following search terms:
(particulate matter OR air pollution OR atmospheric pollution OR environmental pollution OR air pollutants OR atmospheric pollutants OR environmental pollutants) AND (genetic modification OR epigenetic modification) AND (lung diseases OR obstructive lung disease OR obstructive airway disease OR obstructive airways disease OR asthma OR chronic bronchitis OR COPD OR chronic obstructive pulmonary disease OR emphysema OR cardiovascular disease OR heart failure OR coronary artery disease OR congestive heart failure OR congenital heart disease OR peripheral artery disease OR metabolic syndrome), Table 1.

2.4. Inclusion/Exclusion Criteria

We included original articles that examined: i. lung disease; ii. heart disease; iii. epigenetic modification; iv. specific genes/alleles and; v. PM/air pollution. We excluded studies that were: i. reviews/statements; ii. abstracts; iii. no full text available; iv. secondary research on a previous study; v. tobacco/cigarette smoke as a primary source of exposure; vi. prenatal; vii. occupational exposure; viii. heavy metal exposure; ix. lacking mention of a specific allele or loci and; x. not related to genomics/epigenetic modification. Results from the database search were imported into EndNoteX9 (Clarivate Analytics, Philadelphia, PA, USA).

2.5. Data Extraction

In the initial screening, two researchers (Julia Citron (J.C.), Emma Willcocks (E.W.)) screened titles and abstracts for relevant articles. Then, three trained researchers (J.C., E.W., and Anna Nolan (A.N.)) independently reviewed the remaining full-text articles for eligibility. In both screenings, disagreements were resolved through unanimous consensus. Relevant data on genomics, air pollution exposure, and cardiopulmonary disease were compiled to include the specific environmental exposure, cohort characteristics, type of genetic variation or epigenetic modification, and relevant outcomes.

2.6. Methods to Limit Bias

Selection bias was limited by defining fixed inclusion/exclusion criteria and by defining intentional subject matter. Detection bias was addressed by having two individuals screen the search results separately. Reporting bias was limited by using PubMed search filters, screening for peer-reviewed published articles written within 10 years.

3. Results

3.1. Study Selection and Characteristics

The initial PubMed search yielded 95 publications, Figure 1. After screening for studies published within 10 years, 65 papers remained and were assessed for inclusion. During initial title/abstract screening, based on pre-determined criteria, 11 publications were excluded. Exclusion of the remaining full-text articles (n = 54) was based on: i. full-text not available (n = 4); ii. review/statement (n = 23); iii. cigarette/tobacco smoke as a primary source of exposure (n = 8); iv. prenatal studies (n = 4); v. occupational exposure (n = 3); vi. heavy metal exposure (n = 2); vii. no specific allele or loci (n = 1) and; viii. secondary research (n = 2), Figure 1 and Supplemental Tables S1–S3.
The final analysis incorporates seven full-text publications, Table 2: three focused on deoxyribonucleic acid (DNA) methylation and its effects on lung function and pro-inflammatory markers; two focused on oxidative stress after PM exposure and its association with heart defects; and two studied the effects of pollution in populations with genetic variants.

3.2. Effect of Methylation on Lung Function and Inflammation

After air pollution exposure, change in DNA methylation has been correlated with loss of lung function and inflammation [29,32,33]. The work of Sood et al. (2010) used a cohort of smokers with wood smoke exposure to identify that the methylated p16 gene and methylated GATA4 gene were associated with a lower percentage of predicted FEV1 [33]. A methylated GATA4 gene was also associated with a greater chance of airflow obstruction, defined by the Global Initiative for Chronic Obstructive Lung Disease (GOLD) criteria as FEV1/FVC less than 70% [33]. Wood smoke exposure was an overall predictor of COPD, after adjusting for cigarette smoking, and was therefore independently linked with a higher risk of respiratory disease [33].
Somineni et al. (2016) investigated the role of the Ten-Eleven Translocation 1 (TET1) enzyme that regulates DNA methylation in asthma development [29]. A cohort of African American children was found to be at higher risk of developing asthma, after exposure to TRAP when a 5’-C-phosphate-G-3’(CpG) site in the TET1 promoter was methylated [29]. The association between asthma status and cg23602092 methylation was statistically significant (p = 0.040) [29]. This CpG site shows potential as a biomarker for asthma, because cg23602092 methylation was correlated across nasal cells, saliva, and peripheral blood mononuclear cells (PBMC’s) (Pearson’s r ≥ 0.75) [29].
Predisposition to the adverse effects of air pollution exposure was also signified by methylation of LINE-1, Alu, F3, TLR-2, and ICAM-1 in a study of an elderly cohort [32]. Air pollution was linked to changes in markers of coagulation (fibrinogen), inflammation (C-reactive protein), and endothelial function (ICAM-1 and VCAM-1), that may influence risk of cardiovascular disease [32]. There were greater effects of TRAP on fibrinogen and C-reactive protein for those who had either lower methylation of LINE-1 or higher methylation of Alu [32].

3.3. Oxidative Stress Score Associated with Cardiac Deficiency

Oxidative stress at specific loci was another marker of distress after PM exposure, and was correlated with cardiovascular defects [30,34]. QT duration, or the duration of time between the Q and T wave on an electrocardiogram, is a parameter that represents the duration of time for ventricular depolarization and repolarization. Prolongation can lead to fatal arrhythmia and sudden cardiac death. The work of Mordukhovich et al. (2016) described stable associations of QT prolongation with increase of PM2.5 exposure over time in an elderly cohort [30]. The same group found that long-term PM2.5 exposure had the greatest effect on QT, with a 1 year moving average of 9.8µg/m3 of PM2.5 increasing QT duration by as much as 6.3 ms [30]. They found further associations with genetic variants of CAT, GC, GCLM, HMOX-1, and NQO1 genes related to oxidative damage [30].
The oxidative stress response to PM2.5 and black carbon exposure was studied via genetic variants of GSTM1, GSTP1, GSTT1, NQO1, catalase, and HMOX-1 [34]. Black carbon is PM formed through the incomplete combustion of fossil fuels and biomass, and is a major contributor to global climate change. Measurements of systolic blood pressure (SBP) and diastolic blood pressure (DBP) were collected from each participant, to understand the effects of air pollution on cardiac function. Increasing black carbon concentration by 0.43 μg/m3 led SBP to increase by 1.46-mmHg (95% CI: 0.10, 2.82) and DBP to increase by 0.87-mmHg (95% CI: 0.15, 1.59). In contrast, PM2.5 did not have the same effect on SBP or DBP. However, the authors state that the data were limited, because stationary measurements were used to collect data on PM concentrations, which could underestimate PM concentration, whereas longitudinal measures have been demonstrated to more accurately capture variations in everyday exposure [36,37]. Additionally, the relationship between antioxidant-defense-related genetic variants and modifications to blood pressure (BP) after exposure to black carbon or PM2.5 was not statistically significant. However, this is likely attributed to the study’s limitations, as their power to detect a relative effect modification of two was 47% power for a gene prevalence of 20% and 67% power for a gene prevalence of 50% [29].

3.4. Genetic Variation Mediates Effect of Air Pollution

The work of Hwang et al. (2013) looked at the effect of PM2.5 exposure on participants with different alleles of genes in the glutathione-S-transferase (GST) superfamily [31]. Members of the GST superfamily are of interest because they are expressed in the respiratory tract and are related to asthma pathogenesis [38]. Increased risk of asthma after exposure to PM2.5 and O3 was found in allelic variants of val105. Specifically, Ile-105 homozygotes displayed a significant, negative association between risk of asthma and PM2.5 exposure, as well as between asthma and O3 exposure [31]. In addition, Ile-105 homozygotes had a reduced risk of wheezing after PM2.5 and O3 exposure, compared to those with at least one val105 allele [31].
The relationship between genetic composition and air pollution was further studied in a cohort of myocardial infarction (MI) survivors [35]. Genetic variants of fibrinogen and interleukin-6 (IL-6) were of central focus; genomic analysis incorporated single-nucleotide polymorphisms (SNPs) of IL-6, the fibrinogen α-chain gene (FGA), and the β-chain gene (FGB). The amount of IL-6 found in plasma was recorded, to note the levels of inflammatory response correlated with cardiovascular abnormalities [35]. The most significant change was found 6–11 h after carbon monoxide exposure, in those with the IL-6 rs2069832 variant and the FGB rs1800790 variant [35]. There was also a slight change in plasma IL-6 after exposure to NO2, PM2.5, and PM10, but the change was not as clearly defined [35]. There was also a marked difference between carriers of the minor versus major allele types for these expressed polymorphisms [35]. A significant increase in plasma IL-6 levels was found in participants carrying the minor alleles of IL-6 rs2069832 and IL-6 rs2069845, as well as the major allele of IL-6 rs2069840 (p < 0.05) [35].

4. Discussion

This review includes seven articles in which genomics has been identified as an important factor for distinguishing the effects of exposure to air pollution. The application of genomics is promising for its ability to differentiate individuals at greater risk for disease [39,40,41]. The concept of genetic vulnerability has been previously explored, with diseases like breast and ovarian cancer, as well as with type 2 diabetes [42,43,44]. These studies have provided information for early disease detection that can be used to improve patient care and impact patient outcomes.
Genomic analysis is relevant in the field of cardiopulmonary research, and the evaluation of pre-determined genetic or allelic variations within a population has been successful in defining at-risk individuals [31,35]. As described in this review, carriers of different alleles in the GST superfamily had significantly different responses to PM2.5 exposure, and some carriers were at greater risk for developing asthma [31]. Similar findings were seen in those with genetic variants of fibrinogen and IL-6. Different SNPs of IL-6 and fibrinogen resulted in a greater inflammatory response after exposure to air pollution, specifically carbon monoxide, where inflammatory response marked cardiovascular abnormalities [35]. Our group has completed prior analyses on markers of inflammation, including IL-6. Biomarkers of inflammation have been used to identify cases of WTC-LI in a cohort of never smokers who were exposed to WTC-PM exposure [45]. Information on how the genome impacts these pro-inflammatory biomarkers can strengthen our future studies on WTC-PM exposed individuals and the detection of WTC-LI. The ability to detect subpopulations that are especially vulnerable to adverse cardiopulmonary effects provides greater potential to avoid permanent damage, as it allows for quicker implementation of a treatment plan following exposure.
Risk of disease is also affected by external factors, and symptoms can develop after environmental exposures [46,47]. Gene–environment interaction is known to have serious effects on health, and is applicable when discussing exposure-associated cardiopulmonary disease [48,49,50]. Detection of epigenetic modifications that occur after an exposure event has been a key feature of genomics research [51]. Studies have shown how environmental pollutants can alter the presence of DNA methylation [52,53]. Differences in methylation were associated with asthma, COPD, and inflammation [29,32,33]. Specifically, heightened methylation at distinct loci was correlated with low FEV1 function, increased risk of airflow obstruction, and increased risk of childhood asthma [29,33]. A third study reported that decreased methylation at one site was associated with an increase in biomarkers, markers of cardiovascular disease, whereas increased methylation at a separate site was also associated with an increase in these biomarkers [32]. Though there was variation between the amounts of methylation that displayed a significant effect, the study supports the fact that overall level of methylation is a relevant factor in determining exposure susceptibility. DNA methylation regulates expression of genes and is therefore important in gene interactions and subsequent effects on the body [54]. In the studies analyzed, methylation could impact on cardiopulmonary health via this epigenetic mechanism.
Oxidative stress levels are additionally relevant in determining cardiopulmonary health. Reactive oxygen species (ROS) collect within the body and can cause damage to DNA and other cellular structures [55]. Oxidative stress at specific loci was used to identify individuals at risk of disease. Long-term PM2.5 exposure was associated with a prolonged corrected QT duration that was further related to an oxidative stress score, calculated using variants of five oxidative defense genes [30]. Greater genetic vulnerability to oxidative stress leaves subjects at risk of inflammation and can interrupt the autonomic cardiac control that regulates QT duration [30]. However, there was no modification effect of oxidative stress on the association between BP and black carbon exposure [34]. Although the study states that no significant relationship was detected, this may be due to its limited power of detection, and, therefore, more oxidative stress genomics studies are needed before it is possible to conclude its effects on cardiac function.
This review has inherent limitations that were minimized by having clearly defined search criteria. Selection bias was addressed by having pre-determined inclusion/exclusion criteria, in addition to having results screened independently by two individuals. Reporting bias was minimized by selecting for peer-reviewed articles published within the past 10 years on PubMed. There is also bias due to the limited data available on the genomics of exposure-associated cardiopulmonary disease, as genomics is a relatively new field.
This review examines only PM exposure. Although air pollution includes gaseous pollutants that have been linked to cardiopulmonary health, such as ozone, PM is a significant component. Further, although WTC-PM is distinct from ambient PM in its composition and concentration levels, we do not distinctly separate them as PM exposure effects on epigenetics. Nevertheless, the limitations of this study do not affect the clear relevance of genomics research. Analyzing the effects of air pollution exposure through the identification of pre-determined genetic variants, epigenetic modifications, and levels of oxidative stress, has clinical importance. Further research will allow for greater generalization and application. Genomics research has the potential for future impact on PM exposure studies.

5. Conclusions

Genomics research has the potential for significant impact on exposure-associated cardiopulmonary disease. Both the level of methylation and pre-determined genetic variations were shown to significantly mediate the relationship between exposure events and disease state. The effect of oxidative stress on the relationship between exposure events and disease state, in contrast, was inconclusive, indicating that further research is necessary. Nevertheless, there was a consistent, negative impact of environmental exposure on cardiopulmonary health, particularly with regard to COPD, asthma, BP, QT duration, and inflammation.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1660-4601/16/22/4335/s1. Supplemental Table S1: All Search Results; Supplemental Table S2: Search Results After 10 Year Filter; Supplemental Table S3: Full Text Articles that were Reviewed

Author Contributions

All authors made substantial contributions to the study and manuscript preparation. Conceptualization, J.C. and A.N.; Methodology, J.C. and A.N.; Software, J.C., A.N. and G.C.; Validation, J.C., A.N. and E.W.; Formal Analysis, J.C., A.N. and E.W.; Investigation, J.C.; Resources, A.N.; Data Curation, J.C.; Writing—Original Draft Preparation, J.C.; Writing—Review and Editing, J.C., E.W., G.C., S.K. and A.N.; Visualization, J.C. and A.N.; Supervision, A.N.; Project Administration, A.N.; Funding Acquisition, A.N.

Funding

This work was supported, in part, by NHLBI R01HL119326 and CDC/NIOSH U01-OH011300. The funding agencies did not participate in the study design; collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

Acknowledgments

We would like to thank the FDNY rescue workers for their bravery, sacrifice and continued commitment to our work. They continue to be the inspiration for our work.

Conflicts of Interest

The authors declare no conflicts of interest.

Disclaimer

The views expressed in this article do not communicate an official position of the institution, or funding source.

References

  1. Kim, K.H.; Jahan, S.A.; Kabir, E. A review on human health perspective of air pollution with respect to allergies and asthma. Environ. Int. 2013, 59, 41–52. [Google Scholar] [CrossRef]
  2. Zhao, J.; Li, M.; Wang, Z.; Chen, J.; Zhao, J.; Xu, Y.; Wei, X.; Wang, J.; Xie, J. Role of Pm2.5 in the development and progression of copd and its mechanisms. Respir. Res. 2019, 20, 120. [Google Scholar] [CrossRef] [PubMed]
  3. MacNee, W.; Donaldson, A.K. Mechanism of lung injury caused by Pm10 and ultrafine particles with special reference to copd. Eur. Respir. J. Suppl. 2003, 40, 47s–51s. [Google Scholar] [CrossRef] [PubMed]
  4. Gan, W.Q.; FitzGerald, J.M.; Carlsten, C.; Sadatsafavi, M.; Brauer, M. Associations of ambient air pollution with chronic obstructive pulmonary disease hospitalization and mortality. Am. J. Respir. Crit. Care Med. 2013, 187, 721–727. [Google Scholar] [CrossRef] [PubMed]
  5. Soca-Chafre, G.; Hernandez-Pedro, N.; Aviles-Salas, A.; Verson, C.A.; Sanchez, K.C.; Cardona, A.F.; Avila-Moreno, F.; Barrios-Bernal, P.; Flores-Estrada, D.; Arrieta, O. Targeted next generation sequencing identified a high frequency genetic mutated profile in wood smoke exposure-related lung adenocarcinoma patients. Oncotarget 2018, 9, 30499–30512. [Google Scholar] [CrossRef] [PubMed]
  6. Aaron, C.P.; Hoffman, E.A.; Kawut, S.M.; Austin, J.H.M.; Budoff, M.; Michos, E.D.; Stukovsky, K.H.; Sack, C.; Szpiro, A.A.; Watson, K.D.; et al. Ambient air pollution and pulmonary vascular volume on computed tomography: The mesa air pollution and lung cohort studies. Eur. Respir. J. 2019, 53, 1802116. [Google Scholar] [CrossRef]
  7. Sofianopoulou, E.; Kaptoge, S.; Graf, S.; Hadinnapola, C.; Treacy, C.M.; Church, C.; Coghlan, G.; Gibbs, J.S.R.; Haimel, M.; Howard, L.S.; et al. Traffic exposures, air pollution and outcomes in pulmonary arterial hypertension: A Uk cohort study analysis. Eur. Respir. J. 2019, 53, 1801429. [Google Scholar] [CrossRef]
  8. Weiden, M.D.; Kwon, S.; Caraher, E.; Berger, K.I.; Reibman, J.; Rom, W.N.; Prezant, D.J.; Nolan, A. Biomarkers of world trade center particulate matter exposure: Physiology of distal airway and blood biomarkers that predict fev(1) decline. Semin. Respir. Crit. Care Med. 2015, 36, 323–333. [Google Scholar] [CrossRef]
  9. Lippmann, M.; Cohen, M.D.; Chen, L.C. Health effects of world trade center (WTC) dust: An unprecedented disaster’s inadequate risk management. Crit. Rev. Toxicol. 2015, 45, 492–530. [Google Scholar] [CrossRef]
  10. Landrigan, P.J. Health consequences of the 11 september 2001 attacks. Environ. Health Perspect. 2001, 109, A514–A515. [Google Scholar] [CrossRef]
  11. Kwon, S.; Crowley, G.; Caraher, E.J.; Haider, S.H.; Lam, R.; Veerappan, A.; Yang, L.; Liu, M.; Zeig-Owens, R.; Schwartz, T.; et al. Validation of predictive metabolic syndrome biomarkers of world trade center lung injury: A 16-year longitudinal study. Chest 2019, 156, 486–496. [Google Scholar] [CrossRef] [PubMed]
  12. Kwon, S.; Crowley, G.; Mikhail, M.; Lam, R.; Clementi, E.; Zeig-Owens, R.; Schwartz, T.M.; Liu, M.; Prezant, D.J.; Nolan, A. Metabolic syndrome biomarkers of world trade center airway hyperreactivity: A 16-year prospective cohort study. Int. J. Environ. Res. Public Health 2019, 16, 1486. [Google Scholar] [CrossRef] [PubMed]
  13. Caraher, E.J.; Kwon, S.; Haider, S.H.; Crowley, G.; Lee, A.; Ebrahim, M.; Zhang, L.; Chen, L.C.; Gordon, T.; Liu, M.; et al. Receptor for advanced glycation end-products and world trade center particulate induced lung function loss: A case-cohort study and murine model of acute particulate exposure. PLoS ONE 2017, 12, e0184331. [Google Scholar] [CrossRef] [PubMed]
  14. Conesa, A.; Mortazavi, A. The common ground of genomics and systems biology. BMC Syst. Biol. 2014, 8, S1. [Google Scholar] [CrossRef]
  15. van Lanschot, M.C.J.; Bosch, L.J.W.; de Wit, M.; Carvalho, B.; Meijer, G.A. Early detection: The impact of genomics. Virchows Arch. 2017, 471, 165–173. [Google Scholar] [CrossRef]
  16. Worthey, E.A.; Mayer, A.N.; Syverson, G.D.; Helbling, D.; Bonacci, B.B.; Decker, B.; Serpe, J.M.; Dasu, T.; Tschannen, M.R.; Veith, R.L.; et al. Making a definitive diagnosis: Successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet. Med. 2011, 13, 255–262. [Google Scholar] [CrossRef] [Green Version]
  17. Walsh, T.; Shahin, H.; Elkan-Miller, T.; Lee, M.K.; Thornton, A.M.; Roeb, W.; Rayyan, A.A.; Loulus, S.; Avraham, K.B.; King, M.C.; et al. Whole exome sequencing and homozygosity mapping identify mutation in the cell polarity protein Gpsm2 as the cause of nonsyndromic hearing loss Dfnb82. Am. J. Hum. Genet. 2010, 87, 90–94. [Google Scholar] [CrossRef]
  18. Bainbridge, M.N.; Wiszniewski, W.; Murdock, D.R.; Friedman, J.; Gonzaga-Jauregui, C.; Newsham, I.; Reid, J.G.; Fink, J.K.; Morgan, M.B.; Gingras, M.C.; et al. Whole-genome sequencing for optimized patient management. Sci. Transl. Med. 2011, 3, 87re3. [Google Scholar] [CrossRef]
  19. Garcia-Gimenez, J.L.; Seco-Cervera, M.; Tollefsbol, T.O.; Roma-Mateo, C.; Peiro-Chova, L.; Lapunzina, P.; Pallardo, F.V. Epigenetic biomarkers: Current strategies and future challenges for their use in the clinical laboratory. Crit. Rev. Clin. Lab. Sci. 2017, 54, 529–550. [Google Scholar] [CrossRef]
  20. Wu, K.; Li, L.; Thakur, C.; Lu, Y.; Zhang, X.; Yi, Z.; Chen, F. Proteomic characterization of the world trade center dust-activated mdig and c-myc signaling circuit linked to multiple myeloma. Sci. Rep. 2016, 6, 36305. [Google Scholar] [CrossRef]
  21. Cleven, K.L.; Ye, K.; Zeig-Owens, R.; Hena, K.M.; Montagna, C.; Shan, J.; Hosgood, H.D., 3rd; Jaber, N.; Weiden, M.D.; Colbeth, H.L.; et al. Genetic variants associated with fdny Wtc-related sarcoidosis. Int. J. Environ. Res. Public Health 2019, 16, 1830. [Google Scholar] [CrossRef] [PubMed]
  22. Sarapas, C.; Cai, G.; Bierer, L.M.; Golier, J.A.; Galea, S.; Ising, M.; Rein, T.; Schmeidler, J.; Muller-Myhsok, B.; Uhr, M.; et al. Genetic markers for ptsd risk and resilience among survivors of the world trade center attacks. Dis. Markers 2011, 30, 101–110. [Google Scholar] [CrossRef] [PubMed]
  23. Yehuda, R.; Cai, G.; Golier, J.A.; Sarapas, C.; Galea, S.; Ising, M.; Rein, T.; Schmeidler, J.; Muller-Myhsok, B.; Holsboer, F.; et al. Gene expression patterns associated with posttraumatic stress disorder following exposure to the world trade center attacks. Biol. Psychiatry 2009, 66, 708–711. [Google Scholar] [CrossRef] [PubMed]
  24. Banauch, G.I.; Dhala, A.; Alleyne, D.; Alva, R.; Santhyadka, G.; Krasko, A.; Weiden, M.; Kelly, K.J.; Prezant, D.J. Bronchial hyperreactivity and other inhalation lung injuries in rescue/recovery workers after the world trade center collapse. Crit. Care Med. 2005, 33, S102–S106. [Google Scholar] [CrossRef]
  25. Vilcassim, M.J.R.; Thurston, G.D.; Chen, L.C.; Lim, C.C.; Saunders, E.; Yao, Y.; Gordon, T. Exposure to air pollution is associated with adverse cardiopulmonary health effects in international travelers. J. Travel Med. 2019, 26. [Google Scholar] [CrossRef]
  26. Nayebare, S.R.; Aburizaiza, O.S.; Siddique, A.; Carpenter, D.O.; Pope, C.A., 3rd; Mirza, H.M.; Zeb, J.; Aburiziza, A.J.; Khwaja, H.A. Fine particles exposure and cardiopulmonary morbidity in jeddah: A time-series analysis. Sci. Total Environ. 2019, 647, 1314–1322. [Google Scholar] [CrossRef]
  27. Combes, A.; Franchineau, A.G. Fine particle environmental pollution and cardiovascular diseases. Metab. Clin. Exp. 2019, 100S, 153944. [Google Scholar] [CrossRef]
  28. Hu, J.J.; Yu, A.Y.X. Epigenetic response profiles into environmental epigenotoxicant screening and health risk assessment: A critical review. Chemosphere 2019, 226, 259–272. [Google Scholar] [CrossRef]
  29. Somineni, H.K.; Zhang, X.; Myers, J.M.B.; Kovacic, M.B.; Ulm, A.; Jurcak, N.; Ryan, P.H.; Hershey, G.K.K.; Ji, H. Ten-eleven translocation 1 (Tet1) methylation is associated with childhood asthma and traffic-related air pollution. J. Allergy Clin. Immunol. 2016, 137, 797–805. [Google Scholar] [CrossRef]
  30. Mordukhovich, I.; Kloog, I.; Coull, B.; Koutrakis, P.; Vokonas, P.; Schwartz, J. Association between particulate air pollution and qt interval duration in an elderly cohort. Epidemiology 2016, 27, 284–290. [Google Scholar] [CrossRef]
  31. Hwang, B.F.; Young, L.H.; Tsai, C.H.; Tung, K.Y.; Wang, P.C.; Su, M.W.; Lee, Y.L. Fine particle, ozone exposure, and asthma/wheezing: Effect modification by glutathione s-transferase P1 polymorphisms. PLoS ONE 2013, 8, e52715. [Google Scholar] [CrossRef] [PubMed]
  32. Bind, M.A.; Baccarelli, A.; Zanobetti, A.; Tarantini, L.; Suh, H.; Vokonas, P.; Schwartz, J. Air pollution and markers of coagulation, inflammation, and endothelial function: Associations and epigene-environment interactions in an elderly cohort. Epidemiology 2012, 23, 332–340. [Google Scholar] [CrossRef] [PubMed]
  33. Sood, A.; Petersen, H.; Blanchette, C.M.; Meek, P.; Picchi, M.A.; Belinsky, S.A.; Tesfaigzi, Y. Wood smoke exposure and gene promoter methylation are associated with increased risk for copd in smokers. Am. J. Respir. Crit. Care Med. 2010, 182, 1098–1104. [Google Scholar] [CrossRef] [PubMed]
  34. Mordukhovich, I.; Wilker, E.; Suh, H.; Wright, R.; Sparrow, D.; Vokonas, P.S.; Schwartz, J. Black carbon exposure, oxidative stress genes, and blood pressure in a repeated-measures study. Environ. Health Perspect. 2009, 117, 1767–1772. [Google Scholar] [CrossRef]
  35. Ljungman, P.; Bellander, T.; Schneider, A.; Breitner, S.; Forastiere, F.; Hampel, R.; Illig, T.; Jacquemin, B.; Katsouyanni, K.; von Klot, S.; et al. Modification of the interleukin-6 response to air pollution by interleukin-6 and fibrinogen polymorphisms. Environ. Health Perspect. 2009, 117, 1373–1379. [Google Scholar] [CrossRef]
  36. Rojas-Bracho, L.; Suh, H.H.; Koutrakis, P. Relationships among personal, indoor, and outdoor fine and coarse particle concentrations for individuals with copd. J. Expo. Anal. Environ. Epidemiol. 2000, 10, 294–306. [Google Scholar] [CrossRef]
  37. Nethery, E.; Teschke, K.; Brauer, M. Predicting personal exposure of pregnant women to traffic-related air pollutants. Sci. Total Environ. 2008, 395, 11–22. [Google Scholar] [CrossRef]
  38. Korytina, G.F.; Yanbaeva, D.G.; Babenkova, L.I.; Etkina, E.I.; Victorova, T.V. Genetic polymorphisms in the cytochromes P-450 (1a1, 2e1), microsomal epoxide hydrolase and glutathione s-transferase M1, T1, and P1 genes, and their relationship with chronic bronchitis and relapsing pneumonia in children. J. Mol. Med. 2005, 83, 700–710. [Google Scholar] [CrossRef]
  39. Bloss, C.S.; Jeste, D.V.; Schork, N.J. Genomics for disease treatment and prevention. Psychiatr. Clin. North Am. 2011, 34, 147–166. [Google Scholar] [CrossRef]
  40. Kotze, M.J.; Luckhoff, H.K.; Peeters, A.V.; Baatjes, K.; Schoeman, M.; van der Merwe, L.; Grant, K.A.; Fisher, L.R.; van der Merwe, N.; Pretorius, J.; et al. Genomic medicine and risk prediction across the disease spectrum. Crit. Rev. Clin. Lab. Sci. 2015, 52, 120–137. [Google Scholar] [CrossRef]
  41. Ginsburg, G.S.; Willard, A.H.F. Genomic and personalized medicine: Foundations and applications. Transl. Res. 2009, 154, 277–287. [Google Scholar] [CrossRef] [PubMed]
  42. Ford, D.; Easton, D.F.; Bishop, D.T.; Narod, S.A.; Goldgar, D.E. Risks of cancer in brca1-mutation carriers. breast cancer linkage consortium. Lancet 1994, 343, 692–695. [Google Scholar] [CrossRef]
  43. Ali, O. Genetics of type 2 diabetes. World J. Diabetes 2013, 4, 114–123. [Google Scholar] [CrossRef] [PubMed]
  44. Wendt, C.; Margolin, A.S. Identifying breast cancer susceptibility genes-a review of the genetic background in familial breast cancer. Acta Oncol. 2019, 58, 135–146. [Google Scholar] [CrossRef]
  45. Nolan, A.; Naveed, B.; Comfort, A.L.; Ferrier, N.; Hall, C.B.; Kwon, S.; Kasturiarachchi, K.J.; Cohen, H.W.; Zeig-Owens, R.; Glaser, M.S.; et al. Inflammatory biomarkers predict airflow obstruction after exposure to world trade center dust. Chest 2012, 142, 412–418. [Google Scholar] [CrossRef]
  46. Wolff, M.S.; Weston, A.A. Breast cancer risk and environmental exposures. Environ. Health Perspect. 1997, 105, 891–896. [Google Scholar]
  47. Franks, P.W. Gene X environment interactions in type 2 diabetes. Curr. Diabetes Rep. 2011, 11, 552–561. [Google Scholar] [CrossRef]
  48. Smit, L.A.; Bongers, S.I.; Ruven, H.J.; Rijkers, G.T.; Wouters, I.M.; Heederik, D.; Omland, O.; Sigsgaard, T. Atopy and new-onset asthma in young danish farmers and Cd14, Tlr2, and Tlr4 genetic polymorphisms: A nested case-control study. Clin. Exp. Allergy 2007, 37, 1602–1608. [Google Scholar] [CrossRef]
  49. Braun-Fahrlander, C.; Riedler, J.; Herz, U.; Eder, W.; Waser, M.; Grize, L.; Maisch, S.; Carr, D.; Gerlach, F.; Bufe, A.; et al. Environmental exposure to endotoxin and its relation to asthma in school-age children. New Engl. J. Med. 2002, 347, 869–877. [Google Scholar] [CrossRef]
  50. Werner, M.; Topp, R.; Wimmer, K.; Richter, K.; Bischof, W.; Wjst, M.; Heinrich, J. Tlr4 gene variants modify endotoxin effects on asthma. J. Allergy Clin. Immunol. 2003, 112, 323–330. [Google Scholar] [CrossRef]
  51. DeAngelis, J.T.; Farrington, W.J.; Tollefsbol, T.O. An overview of epigenetic assays. Mol. Biotechnol. 2008, 38, 179–183. [Google Scholar] [CrossRef]
  52. Hou, L.; Zhang, X.; Wang, D.; Baccarelli, A. Environmental chemical exposures and human epigenetics. Int. J. Epidemiol. 2012, 41, 79–105. [Google Scholar] [CrossRef]
  53. Terry, M.B.; Ferris, J.S.; Pilsner, R.; Flom, J.D.; Tehranifar, P.; Santella, R.M.; Gamble, M.V.; Susser, E. Genomic DNA methylation among women in a multiethnic New York city birth cohort. Cancer Epidemiol. Biomark. Prev. 2008, 17, 2306–2310. [Google Scholar] [CrossRef]
  54. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef]
  55. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
Figure 1. Study Design and Article Extraction.
Figure 1. Study Design and Article Extraction.
Ijerph 16 04335 g001
Table 1. Search Terms.
Table 1. Search Terms.
PubMed Search(particulate matter OR air pollution OR atmospheric pollution OR environmental pollution OR air pollutants OR atmospheric pollutants OR environmental pollutants) AND (genetic modification OR epigenetic modification) AND (lung diseases OR obstructive lung disease OR obstructive airway disease OR obstructive airways disease OR asthma OR chronic bronchitis OR COPD OR chronic obstructive pulmonary disease OR emphysema OR cardiovascular disease OR heart failure OR coronary artery disease OR congestive heart failure OR congenital heart disease OR peripheral artery disease OR metabolic syndrome)
Abbreviation: COPD: chronic obstructive pulmonary disease.
Table 2. Study Characteristics.
Table 2. Study Characteristics.
First Author [ref]YearEnvironmental ExposureOutcome MeasureResult
Somineni [29]2016TRAPAsthmacg23606092 methylation was associated with increased risk of childhood asthma
Mordukhovich [30]2016PM2.5QT durationHigh allelic risk profiles calculated based on the genetic variants of CAT, GC, GCLM, HMOX-1, and NQO1 were associated with increased QT duration
Hwang [31]2013Sulfur dioxide, nitrogen dioxides, ozone, carbon monoxide and PM2.5Asthma/wheezingIle105 carriers were positively associated with risk of asthma after PM2.5 and O3 exposure
Bind [32]2012Black carbon, carbon monoxide, sulfate, nitrogen dioxide, ozone and PM2.5Inflammatory biomarkersLower methylation of LINE-1 and higher methylation of Alu were both associated with biomarkers of cardiovascular disease
Sood [33]2010Wood smokeCOPDMethylation at the promoter region of p16 predicted lower FEV1 function and methylation at the promoter region of GATA4 was associated with airflow obstruction; wood smoke was an overall predictor of COPD
Mordukhovich [34]2009Black carbon and PM2.5Blood pressureBlack carbon was associated with increased BP and there was no association with antioxidant-defense-related genetic variants
Ljungman [35]2009Carbon monoxide, nitrogen dioxide, PM10 and PM2.5 Inflammatory biomarkersAir pollution has a greater effect on those with SNPs IL-6 rs2069832 and FGB rs1800790
Abbreviations: BP: Blood Pressure; COPD: Chronic Obstructive Pulmonary Disease; FEV1: Forced Expiratory Volume Over One Second; FGB: Fibrinogen β -Chain Gene; HMOX-1: Heme Oxygenase-1; IL-6: Interleukin-6; NQO1: NAD(P)H Quinine Oxidoreductase 1; PM: Particulate Matter; SNP: Single-Nucleotide Polymorphism; TRAP: Traffic-Related Air Pollution.

Share and Cite

MDPI and ACS Style

Citron, J.; Willcocks, E.; Crowley, G.; Kwon, S.; Nolan, A. Genomics of Particulate Matter Exposure Associated Cardiopulmonary Disease: A Narrative Review. Int. J. Environ. Res. Public Health 2019, 16, 4335. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph16224335

AMA Style

Citron J, Willcocks E, Crowley G, Kwon S, Nolan A. Genomics of Particulate Matter Exposure Associated Cardiopulmonary Disease: A Narrative Review. International Journal of Environmental Research and Public Health. 2019; 16(22):4335. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph16224335

Chicago/Turabian Style

Citron, Julia, Emma Willcocks, George Crowley, Sophia Kwon, and Anna Nolan. 2019. "Genomics of Particulate Matter Exposure Associated Cardiopulmonary Disease: A Narrative Review" International Journal of Environmental Research and Public Health 16, no. 22: 4335. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph16224335

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop