Next Article in Journal
Effects of Soil Quality on the Microbial Community Structure of Poorly Evolved Mediterranean Soils
Next Article in Special Issue
A Review of Associations between Externalizing Behaviors and Prenatal Cannabis Exposure: Limitations & Future Directions
Previous Article in Journal
Critical Assessment of Clean-Up Techniques Employed in Simultaneous Analysis of Persistent Organic Pollutants and Polycyclic Aromatic Hydrocarbons in Fatty Samples
Previous Article in Special Issue
Spatially and Temporally Resolved Ambient PM2.5 in Relation to Preterm Birth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prenatal PM2.5 Exposure in Relation to Maternal and Newborn Telomere Length at Delivery

1
Columbia Center for Children’s Environmental Health, New York, NY 10032, USA
2
Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
3
Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
4
Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10128, USA
*
Author to whom correspondence should be addressed.
Submission received: 25 October 2021 / Revised: 16 December 2021 / Accepted: 27 December 2021 / Published: 3 January 2022
(This article belongs to the Special Issue Prenatal Exposures and Children’s Health)

Abstract

:
Particulate matter with an aerodynamic diameter of 2.5 μm or less (PM2.5) is a ubiquitous air pollutant that is increasingly threatening the health of adults and children worldwide. One health impact of elevated PM2.5 exposure is alterations in telomere length (TL)—protective caps on chromosome ends that shorten with each cell division. Few analyses involve prenatal PM2.5 exposure, and paired maternal and cord TL measurements. Here, we analyzed the association between average and trimester-specific prenatal PM2.5 exposure, and maternal and newborn relative leukocyte TL measured at birth among 193 mothers and their newborns enrolled in a New-York-City-based birth cohort. Results indicated an overall negative relationship between prenatal PM2.5 and maternal TL at delivery, with a significant association observed in the second trimester (β = −0.039, 95% CI: −0.074, −0.003). PM2.5 exposure in trimester two was also inversely related to cord TL; however, this result did not reach statistical significance (β = −0.037, 95% CI: −0.114, 0.039), and no clear pattern emerged between PM2.5 and cord TL across the different exposure periods. Our analysis contributes to a limited body of research on ambient air pollution and human telomeres, and emphasizes the need for continued investigation into how PM2.5 exposure during pregnancy influences maternal and newborn health.

1. Introduction

Numerous studies have shown an association between high levels of air pollution and adverse human health outcomes, including in children. In a recent study for the Global Burden of Disease, ambient particulate matter (PM) was listed as the sixth most influential risk factor affecting public health worldwide [1], with fine particles smaller than 2.5 μm in aerodynamic diameter (PM2.5) linked to an estimated 4.2 million deaths globally [2]. Pregnant women represent a particularly high-risk group for the damaging effects of air pollution, given their rapidly changing physiology, including shifts to the immune system and oxidative stress defense system [3]. Recent research also demonstrates that inhaled particles have the potential to cross the placenta, and expose the sensitive and rapidly developing fetus [4]. Prenatal exposure to PM2.5 has been associated with a range of adverse birth outcomes, including preterm birth, reduced birthweight, and stillbirth [5,6,7,8,9], as well as diseases associated with aging [10].
Environmental influences on telomere length (TL) may be one intermediate step linking exposure to air pollutants with adverse health outcomes [11,12]. Telomeres are repetitive, non-coding DNA-protein complexes that function as protective caps on the ends of chromosomes, and contribute to the maintenance of genomic stability and chromosomal integrity, among other functions [13,14,15]. Telomeres shorten with each cell division, and are highly susceptible to oxidative stress due to their guanine-rich structure [16]. Increasing evidence has documented that a range of pro-oxidant environmental exposures, including particulate air pollutants, may be associated with enhanced telomere attrition. For example, in adults, smoking, black carbon, and traffic-related air pollution, as well as PM2.5 exposures have been linked to shorter TL [10,11,12,17]. Longitudinal studies of telomere dynamics in adults have also demonstrated a fixed ranking and tracking of TL across age, suggesting that the early life period may be a critical window for the establishment of inter-individual differences in TL.
In the present study, we extend prior work by investigating maternal exposure to PM2.5 throughout pregnancy in relation to leukocyte TL measured in both cord blood and maternal blood collected at the time of delivery. Our study sample is composed of lower-income, minority women largely recruited from disadvantaged neighborhoods in Northern Manhattan and the South Bronx—communities that are understudied, despite often being subjected to a higher burden of toxic environmental exposures.

2. Materials and Methods

2.1. Sample Studied

The Columbia Center for Children’s Environmental Health (CCCEH) Mothers and Newborns (MN) birth cohort includes 727 Dominican and African American women aged 18–35 recruited between 1998 and 2006 from the prenatal clinics of New York Presbyterian Medical Center, Harlem Hospital, and their affiliated satellite clinics. All women were fluent in English or Spanish, non-smokers, did not use illicit drugs, registered into the study by their 20th week of pregnancy, and were free of diabetes, hypertension, and any known HIV infection. Since being enrolled, the women and children in the MN study have contributed a unique body of data replete with environmental exposures and children’s health outcomes. The current analysis is restricted to a subsample of 193 participants for whom banked maternal and umbilical cord blood samples were available at the time of TL analysis, and residential address during pregnancy was successfully geocoded. All study protocols were approved by the Institutional Review Board of Columbia University. Before each visit, women were informed about all study procedures, and provided written informed consent to participate in their preferred language.

2.2. PM2.5 Exposure

We geocoded maternal residential address during pregnancy as previously described in detail [18,19]. The model that estimates daily residential PM2.5 exposures was previously described in [20]. To assign estimates of fine particulate matter (PM2.5) to individuals in our study, we utilized validated spatio-temporal air pollution exposure models using two sources of air pollution: the New York City Community Air Survey (NYCCAS) data provided directly from the New York City (NYC) Department of Health and Mental Hygiene staff; and regulatory data from the Environmental Protection Agency’s Air Quality System (AQS) (https://aqsdr1.epa.gov/aqsweb/aqstmp/airdata/download_files.html, accessed on 16 December 2021). NYCCAS data were collected at 60–150 locations. Daily measurements were estimated by averaging the measurements corresponding to the 2 weeks surrounding each day. To capture true daily air quality variation in the models, we also included daily data from the NYC Department of Environmental Conservation’s regulatory monitors, which collect data on an everyday or every-third-day schedule. Detailed temporal patterns were captured using data from the EPA regulatory monitors, which collect data on an every-day or every-third-day schedule. Residential PM2.5 exposures were predicted using mother’s residence at the time of delivery. Information on residential relocation, commuting patterns, or time-activity behaviors shaping individual exposures during pregnancy was not available; thus, there is a potential for misclassification of exposure for a small number of participants. Notably, in 2001, PM2.5 measurements were collected every 3 days instead of daily, resulting in fewer data points available to calculate average PM2.5 for each of the three trimesters than for the entire pregnancy in 2001. For example, if a sample was missing data from trimester one, but did have data from trimesters two and three, entire pregnancy data values could still be calculated through mean across time using available values.

2.3. Telomere Length Measurement

Umbilical cord blood and maternal blood were collected at time of delivery by research workers at the CCCEH. Maternal blood was drawn by venipuncture, and all blood samples were collected using green-top tubes lined with sodium heparin to reduce coagulation. Samples were centrifuged in order to separate buffy coat containing blood leukocytes from whole blood, and subsequently processed and stored at −80 °C in the CCCEH laboratory. Genomic DNA (100–500 ng) from 197 mother-newborn pairs was separated from leukocytes using an identical standard phenol-chloroform protocol. A Nanodrop 1000 spectrophotometer (ThermoFisher Scientific, Waltham, MA, USA) was used to confirm the purity of the cord blood DNA with an applied 260/280 ratio. Telomere repeat sequence copy number (T) relative to single gene (S: albumin) copy number was measured using multiplex monochrome quantitative polymerase chain reaction (MMqPCR) on a Bio-Rad CFX96 lightcycler (Bio-Rad Laboratories, Hercules, CA, USA) at the Columbia University Laboratory of Precision Environmental Health [21]. Information pertaining to the assay has been previously described in detail following the recently developed minimum reporting recommendations for qPCR TL measurement established by the Telomere Research Network [22,23]. Relative leukocyte telomere length, hereafter rLTL, measured by qPCR, reflects the average TL across all chromosomes for all cells sampled.

2.4. Statistical Analysis

We examined descriptive statistics for each covariate, cord and maternal rLTL at delivery, and PM2.5 exposures averaged across each trimester, as well as the total pregnancy period. For each trimester and the entire pregnancy, we used multivariable linear regression to examine the association between PM2.5 dichotomized at the median, and maternal or cord blood rLTL in a separate model. We also analyzed the association between continuous PM2.5 and maternal and cord rLTL. Equation (1) shows the formula for the multivariable linear regression model used to test the association between maternal rLTL and dichotomized PM2.5 in each trimester and total pregnancy, with each iteration of the equation incorporating only adjustments to the outcome or main predictor. “High” indicates higher than the median. The first trimester is shown as an example. Equation (2) shows the formula used for analysis of continuous PM2.5 in each trimester and total pregnancy. All estimates, confidence intervals, and p-values of β 1 are reported in the Section 3. Results with corresponding alpha values of less than 0.05 were considered significant.
E ( maternal   rLTL   |   PM 2.5 ,   covariates ) = β 0 + β 1 𝕀 ( PM 2.5 T 1 = high ) + + β k covariate k
E ( maternal   rLTL   |   PM 2.5 ,   covariates ) = β 0 + β 1 PM 2.5 T 1 + + β k covariate k

2.5. Covariates

We examined the relationship between prenatal PM2.5 exposure and maternal and cord rLTL, adjusting for several sociodemographic and obstetric characteristics, including: gestational age at birth, child sex (male vs. female), ethnicity (Dominican vs. African American), maternal age (continuous in years), and maternal education (<high school vs. high school or more). Because ambient PM2.5 varies by season, with higher levels being recorded during winter and summer months, we adjusted for season of conception [24]. Storage time of blood samples, and a squared term of storage time were also included to account for their nonlinear influence on rLTL. Because maternal rLTL is a strong determinant of newborn rLTL, we ran a second newborn model, also including maternal rLTL as a covariate in order to uncover the unique effects of PM2.5 on newborn rLTL. We acknowledge that smoking is a known predictor of telomere length; however, because one of the inclusion criteria for this cohort was that mothers were non-smoking, we did not include smoking as a covariate.

3. Results

Out of the 727 women enrolled in the MN Study, 197 mother–newborn pairs had available data on rLTL. Of these, 193 had residential address information that was successfully geocoded, allowing for the generation of PM2.5 estimates for each trimester and across total pregnancy. All women self-identified as either Dominican (63%) or African American (37%), and lived in Northern Manhattan or the South Bronx at the time of recruitment. At delivery, mothers were on average 25 years old, and 37% had less than a high school degree or equivalent. The average gestational age for newborns was 39 weeks, which is expected, as preterm births were largely excluded by design. Table 1 provides additional sociodemographic and birth characteristics of the study participants. It also includes medians, and first and third quartiles for PM2.5 exposure data for all three trimesters and average pregnancy, as well as summary statistics for umbilical cord and maternal rLTL. No sociodemographic or lifestyle characteristics significantly differed between the 193 participants included in the analytic sample, and those enrolled in the cohort, but excluded from the present analysis (Supplemental Table S1). Total pregnancy (median: 17.0 µg/m3 vs. 16.5 µg/m3) and first trimester (17.4 µg/m3 vs. 16.7 µg/m3) PM2.5 levels were slightly higher among excluded participants compared to included participants, which reflects that excluded participants were more likely to be enrolled earlier during the recruitment period when air pollution levels in New York City were higher; participants enrolled earlier during the study were less likely to be included, as they were less likely to have biobanked blood samples remaining at the time of this study.
As expected, we observed a significantly positive correlation between cord blood rLTL and maternal rLTL (Pearson’s correlation r = 0.25 with p < 0.001). The results of regression analyses examining PM2.5 in relation to rLTL are presented in Table 2, and a correlation plot is depicted in Figure 1. PM2.5 exposure during pregnancy was inversely associated with maternal rLTL at delivery, with a significant association observed for exposure occurring during the second trimester (β = −0.039, 95% CI: −0.074, −0.003). Likewise, PM2.5 exposure during the second trimester was inversely associated with cord blood rLTL with (β = −0.017, 95% CI: −0.092, 0.058) and without (β = −0.037, 95% CI: −0.114, 0.039) adjustment for maternal rLTL; however, this finding did not reach statistical significance. For exposure occurring during the first and third trimesters, as well as the pregnancy average, we observed positive, albeit not statistically significant, associations with cord blood rLTL regardless of adjustment for maternal rLTL, indicating generally null results, and no clear pattern of association across the exposure periods.

4. Discussion

In this study, we found that exposure to PM2.5 during pregnancy was inversely associated with maternal rLTL at delivery, with the strongest associations identified for exposure occurring during the second trimester (Table 2). In contrast, no clear pattern or significant associations were observed with cord rLTL.
Research supports that the maintenance of TL differs between life stages [13,22,25,26], suggesting that vulnerability to exposures with the potential to alter TL may vary between newborns and adults. This may help explain our observed differences in results between maternal and cord TL. Though it is critical to study changes in TL in newborns due to their developmental susceptibility, it is equally important to analyze TL changes in adults, given the biological consequences of shortened TL in adulthood, among them, shortened lifespan and age-related diseases [27]. Furthermore, pregnant women specifically might be at greater risk of TL shortening than the rest of the adult population because of biological changes during pregnancy, including substantial changes to vascular physiology, metabolism, reproductive organs, endocrine activity, and the immune system [3].
To our knowledge, only two prior studies have examined PM2.5 in relation to TL among pregnant women [28,29]. Among a sample of 199 healthy women in Italy, PM2.5 exposure during early pregnancy (range = 7 to 69 μg/m3, all medians < 30) was not associated with leukocyte TL towards the end of the first trimester [28]. Unfortunately, other exposure and outcome periods were not considered, making it difficult to directly compare with our findings. Similarly, among a sample of 296 women in Denmark, PM2.5 exposure (mean ± SD: 11.5 μg/m3) was not associated with maternal leukocyte TL at delivery when averaged over pregnancy or by trimester [29]. Variation in levels of ambient air pollution in New York City, compared to Milan, Italy, and Copenhagen, Denmark, may help explain the difference in results between our study and those in Europe.
In addition to the limited research examining PM2.5 in relation to maternal TL during pregnancy or at delivery, five recent studies have examined PM2.5 in relation to cord blood TL with mixed results. Consistent with our finding of an inverse, albeit insignificant, association with exposure occurring during the second trimester, two studies based in the northeast U.S. (n = 155, mean PM2.5 8.3 µg/m3) and in Belgium (n = 641, mean PM2.5 = 13.4 µg/m3) identified inverse associations between PM2.5 and cord blood TL, with exposure occurring during the second trimester only [10,30]. The study in Belgium additionally found a positive association with exposure during the third trimester [10]. Other studies based in Copenhagen (n = 296, mean PM2.5 = 11.5 µg/m3), Mexico City (n = 423, mean PM2.5 = 22.8 µg/m3), and Wuhan (n = 743, mean PM2.5 = 69.1 µg/m3) have reported a range of associations between PM2.5 and cord blood TL, including inverse relations with exposure occurring during the first [31] and third trimesters [29,32], and positive relations with exposure during the second [29,31] and third trimesters [31]. Positive associations between cord TL and PM2.5 exposure, as we observed during the 1st and 3rd trimesters, have contributed to some researchers purporting a compensatory or overcompensatory mechanism in which the biology of the fetus attempts to balance out the adverse effects of earlier exposures in gestation [10,31].
Taken together, these results do not support the identification of a specific exposure period for heightened telomere susceptibility, but rather paint a complicated picture that likely reflects differences between study designs and a shifting telomere dynamic across the course of pregnancy. Notably, pregnancy is characterized by an enormous number of cell divisions. For example, the mother experiences expanded blood volume [33,34,35] and changes to the immune system that enable support of the semi-allogenic fetus [36], both of which may influence leukocyte TL. These changes, typical of pregnancy, present a challenge for studies examining TL in relation to time-varying environmental exposures, such as PM2.5, and likely contribute to the inconsistent windows of susceptibility identified by studies conducted to date.
Telomerase, the reverse transcriptase that elongates telomeres in stem and other replicative cells, is difficult to measure, and this difficulty further complicates our understanding of how PM2.5 is related to TL during pregnancy. Though the mature oocyte does not display telomerase activity, soon after fertilization, and throughout gestation, telomerase levels increase, allowing telomeres to be maintained across the embryonic and fetal developmental periods, despite the enormous cellular proliferation that occurs in utero [37]. Though speculative, it is plausible that PM2.5 affects fetal telomeres during periods of down-regulated telomerase activity, or alternatively acts directly on telomerase. However, we acknowledge that whether telomerase is naturally downregulated during the second trimester or a target of PM2.5 remains unknown. The greater activity of telomerase in fetal compared to adult tissue may also, in part, explain our finding of greater inverse associations between PM2.5 and maternal compared to cord blood rLTL. The mother is also more directly exposed to PM2.5, with the placenta likely protecting the fetus from at least some particulate exposure. However, we note that beginning during the second trimester, the placental barrier begins to thin, potentially enhancing maternal to fetal particle exchange [10]. Future research investigating telomere dynamics across gestation in greater detail may help uncover relevant biological mechanisms, and provide insight on susceptible windows of exposure.
A key strength of the current study is our usage of paired maternal and cord TL measurements, as there is only one other study analyzing prenatal PM2.5 exposure with this feature [29]. Though our sample size is relatively small, our study is strengthened by its thorough evaluation of possible confounding variables; however, we acknowledge there may have been additional important covariates, such as paternal characteristics, that we were unable to control for. Additionally, our results may have been affected by the contamination of cord blood from maternal blood, a documented phenomenon [38]. Our comprehensive estimates of daily PM2.5 exposure provide a detailed reflection of each study participant’s individual exposure in all three trimesters of pregnancy. However, we recognize that because our estimates were based on maternal residence outdoors, they could not account for other exposure sources away from the home, such as a daily commute to work, or air quality differences inside the home [39] that could also contribute to a person’s overall PM2.5 exposure profile. Additionally, as previously stated, no information on residential moves during pregnancy was available, and we understand potential differences in exposure between addresses may have influenced our results. However, prior research suggests that most residential moves during pregnancy are relatively short distances, and thus, should not influence PM2.5 estimates to a great degree [40]. In a previous publication, we discussed the potential impact of long-term storage on human telomere length [22]. Because of the longitudinal study design, maternal and cord blood samples from dyads enrolled into the cohort the earliest had been stored for 18.5 years. Our results showed that storage time was not a predictor of maternal TL, but was strongly negatively correlated with cord telomere length at each PM2.5 measurement period (individual trimesters and averaged total pregnancy). Thus, our borderline significant association between lengthened cord TL and total pregnancy PM2.5 exposure may have been affected by storage-related alterations. We also acknowledge that PM2.5 is only one component of the complex mixture of ambient air pollutants that humans are exposed to [41,42,43]. Future research that is able to more comprehensively exam the mixture of joint exposures may provide additional insight on how air pollution affects TL. Lastly, though scientific research involving underrepresented minority groups, such as the present study, is highly valuable, we acknowledge that the results of our study may not be generalizable to a broader population.

5. Conclusions

In this analysis, we found that PM2.5 exposure during pregnancy was associated with significantly shorter maternal rLTL at the time of delivery; however, no clear associations emerged when examining cord blood rLTL. Our study involving corresponding cord and maternal TL contributes to the limited body of research on human telomeres and PM2.5, an environmental pollutant that increasingly threatens human health worldwide.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/toxics10010013/s1, Table S1: Characteristics of excluded participants (N = 534).

Author Contributions

Conceptualization, T.D., K.W.R., W.C., J.G., J.B.H., F.P. and S.W.; Methodology, T.D., K.W.R., W.C., J.G., J.B.H., F.P. and S.W.; Software, S.W. and J.G.; Validation, J.B.H., F.P. and S.W.; Formal Analysis, J.G. and S.W.; Investigation, J.B.H. and F.P.; Resources, D.T.; Data Curation, J.B.H., J.G. and S.W.; Writing—Original Draft Preparation, T.D.; Writing—Review and Editing, T.D., J.G., W.C., S.W., F.P., K.W.R., J.B.H. and D.T.; Visualization, T.D. and J.G.; Supervision, J.B.H. and F.P.; Project Administration, J.B.H., F.P. and K.W.R.; Funding Acquisition, J.B.H. and F.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Blanchette Hooker Rockefeller Fund; NIH Office of the Director [UG3OD023290]; NIH [R03ES026416]; NIH [P30ES009089]; National Institute of Environmental Health Sciences [P50ES09600]; National Institute of Environmental Health Sciences [R01ES13163]; National Institute of Environmental Health Sciences [R01ES014393]; National Institute of Environmental Health Sciences [P01ES09600]; National Institute of Environmental Health Sciences [R01ES08977]; New York Community Trust; John and Wendy Neu Foundation; U.S. Environmental Protection Agency [RD83615401]; U.S. Environmental Protection Agency [RD832141]; U.S. Environmental Protection Agency [RD82702701]; U.S. Environmental Protection Agency [RD834509].

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Columbia University (AAAA6110, Renewal Approval: 26 June 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

We will share all published resources generated with NIH funds with interested investigators. After the acceptance for publication of the main findings from the final data set, the study data, free of identifiers, will be shared, under the auspices of the study’s MPI, with other researchers, to the extent allowed by the Institutional Review Board. The Columbia Children Center for Environmental Health (CCCEH) Data Management Core (DMC) will prepare data files for analysis following NIH guidelines and the existing CCCEH Data Use policy. All requests for data must be in writing, for documentation, and addressed to Herbstman.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Forouzanfar, M.H.; GBD 2015 Risk Factors Collaborators. Global, regional, and national comparative risk assessment of 79 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1659–1724. [Google Scholar] [CrossRef] [Green Version]
  2. Cohen, A.J.; Brauer, M.; Burnett, R.; Anderson, H.R.; Frostad, J.; Estep, K.; Balakrishnan, K.; Brunekreef, B.; Dandona, L.; Dandona, R.; et al. Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: An analysis of data from the Global Burden of Diseases Study 2015. Lancet 2017, 389, 1907–1918. [Google Scholar] [CrossRef] [Green Version]
  3. Varshavsky, J.; Smith, A.; Wang, A.; Hom, E.; Izano, M.; Huang, H.; Padula, A.; Woodruff, T. Heightened susceptibility: A review of how pregnancy and chemical exposures influence maternal health. Reprod. Toxicol. 2020, 92, 14–56. [Google Scholar] [CrossRef]
  4. Bové, H.; Bongaerts, E.; Slenders, E.; Bijnens, E.M.; Saenen, N.D.; Gyselaers, W.; Van Eyken, P.; Plusquin, M.; Roeffaers, M.B.J.; Ameloot, M.; et al. Ambient black carbon particles reach the fetal side of human placenta. Nat. Commun. 2019, 10, 3866. [Google Scholar] [CrossRef]
  5. Clemens, T.; Turner, S.; Dibben, C. Maternal exposure to ambient air pollution and fetal growth in North-East Scotland: A population-based study using routine ultrasound scans. Environ. Int. 2017, 107, 216–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Pedersen, M.; Giorgis-Allemand, L.; Bernard, C.; Aguilera, I.; Andersen, A.-M.N.; Ballester, F.; Beelen, R.M.J.; Chatzi, L.; Cirach, M.; Danilevičiūtė, A.; et al. Ambient air pollution and low birthweight: A European cohort study (ESCAPE). Lancet Respir. Med. 2013, 1, 695–704. [Google Scholar] [CrossRef]
  7. Poirier, A.; Dodds, L.; Dummer, T.; Rainham, D.; Maguire, B.; Johnson, M. Maternal Exposure to Air Pollution and Adverse Birth Outcomes in Halifax, Nova Scotia. J. Occup. Environ. Med. 2015, 57, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
  8. Qian, Z.; Liang, S.; Yang, S.; Trevathan, E.; Huang, Z.; Yang, R.; Wang, J.; Hu, K.; Zhang, Y.; Vaughn, M.; et al. Ambient air pollution and preterm birth: A prospective birth cohort study in Wuhan, China. Int. J. Hyg. Environ. Health 2015, 219, 195–203. [Google Scholar] [CrossRef] [PubMed]
  9. Siddika, N.; Balogun, H.A.; Amegah, A.K.; Jaakkola, J.J.K. Prenatal ambient air pollution exposure and the risk of stillbirth: Systematic review and meta-analysis of the empirical evidence. Occup. Environ. Med. 2016, 73, 573–581. [Google Scholar] [CrossRef] [PubMed]
  10. Martens, D.S.; Cox, B.; Janssen, B.G.; Clemente, D.B.P.; Gasparrini, A.; Vanpoucke, C.; Lefebvre, W.; Roels, H.A.; Plusquin, M.; Nawrot, T.S. Prenatal Air Pollution and Newborns’ Predisposition to Accelerated Biological Aging. JAMA Pediatr. 2017, 171, 1160–1167. [Google Scholar] [CrossRef]
  11. De Felice, B.; Nappi, C.; Zizolfi, B.; Guida, M.; Sardo, A.D.S.; Bifulco, G.; Guida, M. Telomere shortening in women resident close to waste landfill sites. Gene 2012, 500, 101–106. [Google Scholar] [CrossRef]
  12. Hoxha, M.; Dioni, L.; Bonzini, M.; Pesatori, A.C.; Fustinoni, S.; Cavallo, D.; Carugno, M.; Albetti, B.; Marinelli, B.; Schwartz, J.; et al. Association between leukocyte telomere shortening and exposure to traffic pollution: A cross-sectional study on traffic officers and indoor office workers. Environ. Health 2009, 8, 41. [Google Scholar] [CrossRef] [Green Version]
  13. Perera, F.; Lin, C.-J.; Qu, L.; Tang, D. Shorter telomere length in cord blood associated with prenatal air pollution exposure: Benefits of intervention. Environ. Int. 2018, 113, 335–340. [Google Scholar] [CrossRef] [PubMed]
  14. Wei, H.; Zhang, C.; Silveyra, P. The Relationships Between Prenatal Smoking Exposure and Telomere Lengths in Fetuses, Infants, and Children: A Systematic Literature Review. UJAN 2020, 31, 243–252. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, M.; Wang, L.; Song, L.; Liu, B.; Liu, Y.; Bi, J.; Liu, Q.; Chen, K.; Li, Y.; Xia, W.; et al. The association between prenatal exposure to thallium and shortened telomere length of newborns. Chemosphere 2021, 265, 129025. [Google Scholar] [CrossRef] [PubMed]
  16. Coluzzi, E.; Colamartino, M.; Cozzi, R.; Leone, S.; Meneghini, C.; O’Callaghan, N.; Sgura, A. Oxidative Stress Induces Persistent Telomeric DNA Damage Responsible for Nuclear Morphology Change in Mammalian Cells. PLoS ONE 2014, 9, e110963. [Google Scholar] [CrossRef]
  17. McCracken, J.; Baccarelli, A.; Hoxha, M.; Dioni, L.; Melly, S.; Coull, B.; Suh, H.; Vokonas, P.; Schwartz, J. Annual Ambient Black Carbon Associated with Shorter Telomeres in Elderly Men: Veterans Affairs Normative Aging Study. Environ. Health Perspect. 2010, 118, 1564–1570. [Google Scholar] [CrossRef] [Green Version]
  18. Huang, Y.; Kioumourtzoglou, M.-A.; Mittleman, M.A.; Ross, Z.; Williams, M.A.; Friedman, A.M.; Schwartz, J.; Wapner, R.J.; Ananth, C.V. Air Pollution and Risk of Placental Abruption: A Study of Births in New York City, 2008–2014. Am. J. Epidemiol. 2021, 190, 1021–1033. [Google Scholar] [CrossRef]
  19. Wang, Y.; Perera, F.; Guo, J.; Riley, K.W.; Durham, T.; Ross, Z.; Ananth, C.V.; Baccarelli, A.; Wang, S.; Herbstman, J.B. A methodological pipeline to generate an epigenetic marker of prenatal exposure to air pollution indicators. Epigenetics 2021, 1–9. [Google Scholar] [CrossRef]
  20. Ross, Z.; Ito, K.; Johnson, S.; Yee, M.; Pezeshki, G.; Clougherty, J.E.; Savitz, D.; Matte, T. Spatial and temporal estimation of air pollutants in New York City: Exposure assignment for use in a birth outcomes study. Environ. Health 2013, 12, 51. [Google Scholar] [CrossRef] [Green Version]
  21. Cawthon, R.M. Telomere length measurement by a novel monochrome multiplex quantitative PCR method. Nucleic Acids Res. 2009, 37, e21. [Google Scholar] [CrossRef] [Green Version]
  22. Cowell, W.; Tang, D.; Yu, J.; Guo, J.; Wang, S.; Baccarelli, A.A.; Perera, F.; Herbstman, J.B. Telomere dynamics across the early life course: Findings from a longitudinal study in children. Psychoneuroendocrinology 2021, 129, 105270. [Google Scholar] [CrossRef]
  23. Lindrose, A.; Drury, S. Minimum Reporting Recommendations for PCR-based Telomere Length Measurement. Open Sci. Framew. 2020. [Google Scholar] [CrossRef]
  24. Zhao, X.; Zhang, X.; Xu, X.; Xu, J.; Meng, W.; Pu, W. Seasonal and diurnal variations of ambient PM2.5 concentration in urban and rural environments in Beijing. Atmos. Environ. 2009, 43, 2893–2900. [Google Scholar] [CrossRef]
  25. Factor-Litvak, P.; Susser, E.; Aviv, A. Environmental Exposures, Telomere Length at Birth, and Disease Susceptibility in Later Life. JAMA Pediatr. 2017, 171, 1143. [Google Scholar] [CrossRef]
  26. Marchetto, N.M.; Glynn, R.; Ferry, M.L.; Ostojic, M.; Wolff, S.M.; Yao, R.; Haussmann, M.F. Prenatal stress and newborn telomere length. Am. J. Obstet. Gynecol. 2016, 215, 94.e1–94.e8. [Google Scholar] [CrossRef] [Green Version]
  27. Liu, B.; Song, L.; Zhang, L.; Wu, M.; Wang, L.; Cao, Z.; Xiong, C.; Zhang, B.; Li, Y.; Xia, W.; et al. Prenatal second-hand smoke exposure and newborn telomere length. Pediatr. Res. 2020, 87, 1081–1085. [Google Scholar] [CrossRef] [PubMed]
  28. Iodice, S.; Hoxha, M.; Ferrari, L.; Carbone, I.F.; Anceschi, C.; Miragoli, M.; Pesatori, A.C.; Persico, N.; Bollati, V. Particulate Air Pollution, Blood Mitochondrial DNA Copy Number, and Telomere Length in Mothers in the First Trimester of Pregnancy: Effects on Fetal Growth. Oxidative Med. Cell. Longev. 2018, 2018, 5162905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Scholten, R.H.; Møller, P.; Andersen, Z.J.; Dehlendorff, C.; Khan, J.; Brandt, J.; Ketzel, M.; Knudsen, L.E.; Mathiesen, L. Telomere length in newborns is associated with exposure to low levels of air pollution during pregnancy. Environ. Int. 2021, 146, 106202. [Google Scholar] [CrossRef]
  30. Lee, A.G.; Cowell, W.; Kannan, S.; Ganguri, H.B.; Nentin, F.; Wilson, A.; Coull, B.A.; Wright, R.O.; Baccarelli, A.; Bollati, V.; et al. Prenatal particulate air pollution and newborn telomere length: Effect modification by maternal antioxidant intakes and infant sex. Environ. Res. 2020, 187, 109707. [Google Scholar] [CrossRef] [PubMed]
  31. Rosa, M.J.; Hsu, H.-H.L.; Just, A.C.; Brennan, K.J.; Bloomquist, T.; Kloog, I.; Pantic, I.; García, A.M.; Wilson, A.; Coull, B.A.; et al. Association between prenatal particulate air pollution exposure and telomere length in cord blood: Effect modification by fetal sex. Environ. Res. 2019, 172, 495–501. [Google Scholar] [CrossRef] [PubMed]
  32. Song, L.; Zhang, B.; Liu, B.; Wu, M.; Zhang, L.; Wang, L.; Xu, S.; Cao, Z.; Wang, Y. Effects of maternal exposure to ambient air pollution on newborn telomere length. Environ. Int. 2019, 128, 254–260. [Google Scholar] [CrossRef]
  33. Costantine, M.M. Physiologic and pharmacokinetic changes in pregnancy. Front. Pharmacol. 2014, 5, 65. [Google Scholar] [CrossRef]
  34. Pritchard, J.A. Changes in the Blood Volume During Pregnancy and Delivery. Anesthesiology 1965, 26, 393–399. [Google Scholar] [CrossRef] [PubMed]
  35. Vricella, L.K. Emerging understanding and measurement of plasma volume expansion in pregnancy. Am. J. Clin. Nutr. 2017, 106, 1620S–1625S. [Google Scholar] [CrossRef]
  36. Koch, C.A.; Platt, J.L. T cell recognition and immunity in the fetus and mother. Cell. Immunol. 2007, 248, 12–17. [Google Scholar] [CrossRef] [Green Version]
  37. Eisenberg, D.T.A. An evolutionary review of human telomere biology: The thrifty telomere hypothesis and notes on potential adaptive paternal effects. Am. J. Hum. Biol. 2011, 23, 149–167. [Google Scholar] [CrossRef] [PubMed]
  38. Morin, A.M.; Gatev, E.; McEwen, L.M.; MacIsaac, J.L.; Lin, D.T.S.; Koen, N.; Czamara, D.; Räikkönen, K.; Zar, H.J.; Koenen, K.; et al. Maternal blood contamination of collected cord blood can be identified using DNA methylation at three CpGs. Clin. Epigenet. 2017, 9, 75. [Google Scholar] [CrossRef]
  39. Lin, N.; Mu, X.; Wang, G.; Ren, Y.; Su, S.; Li, Z.; Wang, B.; Tao, S. Accumulative effects of indoor air pollution exposure on leukocyte telomere length among non-smokers. Environ. Pollut. 2017, 227, 1–7. [Google Scholar] [CrossRef] [PubMed]
  40. Lupo, P.; Symanski, E.; Chan, W.; Mitchell, L.E.; Waller, D.; Canfield, M.A.; Langlois, P.H. Differences in exposure assignment between conception and delivery: The impact of maternal mobility. Paediatr. Perinat. Epidemiol. 2010, 24, 200–208. [Google Scholar] [CrossRef]
  41. Winquist, A.; Kirrane, E.; Klein, M.; Strickland, M.; Darrow, L.A.; Sarnat, S.E.; Gass, K.; Mulholland, J.; Russell, A.; Tolbert, P. Joint Effects of Ambient Air Pollutants on Pediatric Asthma Emergency Department Visits in Atlanta, 1998–2004. Epidemiology 2014, 25, 666–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Dominici, F.; Peng, R.D.; Barr, C.D.; Bell, M. Protecting Human Health From Air Pollution: Shifting From a Single-pollutant to a Multipollutant Approach. Epidemiology 2010, 21, 187–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Billionnet, C.; Sherrill, D.; Annesi-Maesano, I. Estimating the Health Effects of Exposure to Multi-Pollutant Mixture. Ann. Epidemiol. 2012, 22, 126–141. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Correlation analysis of maternal rLTL, cord blood rLTL and PM2.5 exposures averaged across each trimester, as well as PM2.5 exposures at the total pregnancy period. A blank represents an insignificant p-value at the significance level 0.05.
Figure 1. Correlation analysis of maternal rLTL, cord blood rLTL and PM2.5 exposures averaged across each trimester, as well as PM2.5 exposures at the total pregnancy period. A blank represents an insignificant p-value at the significance level 0.05.
Toxics 10 00013 g001
Table 1. Characteristics and cord and maternal relative leukocyte telomere length (rLTL) in the CCCEH Mothers and Newborns birth cohort (n = 193).
Table 1. Characteristics and cord and maternal relative leukocyte telomere length (rLTL) in the CCCEH Mothers and Newborns birth cohort (n = 193).
Mean ± SD or N (%)
African American72 (37.3)
Dominican121 (62.7)
<High school education or equivalent72 (37.3)
Child sex (girl)111 (57.5)
Season of conception
Spring48 (24.9)
Summer43 (22.3)
Fall44 (22.8)
Winter58 (30.1)
Maternal age (years)25.43 ± 5.18
Gestational age (weeks)39.39 ± 1.30
Maternal rLTL at delivery1.02 ± 0.13
Umbilical cord rLTL1.22 ± 0.26
PM2.5 (µg/m3), median (Q1, Q3)
Total pregnancy16.53 (15.69, 17.71)
First trimester16.68 (15.25, 18.76)
Second trimester16.59 (14.67, 18.31)
Third trimester16.78 (14.97, 18.48)
Table 2. Associations between binary (high versus low) PM2.5 exposure a during pregnancy and rLTL in cord blood and maternal blood collected at delivery (n = 193).
Table 2. Associations between binary (high versus low) PM2.5 exposure a during pregnancy and rLTL in cord blood and maternal blood collected at delivery (n = 193).
Cord rLTLCord Rltl bMaternal rLTL
PM2.5 Exposureβ (95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p-Value
Total pregnancy0.063 (−0.021, 0.147)0.1410.083 (0.001, 0.164)0.047−0.034 (−0.074, 0.005)0.087
1st trimester0.039 (−0.039, 0.117)0.3230.051 (−0.024, 0.127)0.183−0.022 (−0.059, 0.015)0.240
2nd trimester−0.037 (−0.114, 0.039)0.337−0.017 (−0.092, 0.058)0.657−0.039 (−0.074, −0.003)0.034
3rd trimester0.042 (−0.036, 0.120)0.2920.039 (−0.037, 0.114)0.3130.006 (−0.031, 0.042)0.765
Abbreviations: PM2.5: fine particulate matter; rLTL: relative leukocyte telomere length. Adjusted for: sample storage time, sample storage time squared, gestational age, conception season, child sex, ethnicity, maternal age, and maternal education. a Dichotomized at each median. b Additionally adjusted for maternal rLTL.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Durham, T.; Guo, J.; Cowell, W.; Riley, K.W.; Wang, S.; Tang, D.; Perera, F.; Herbstman, J.B. Prenatal PM2.5 Exposure in Relation to Maternal and Newborn Telomere Length at Delivery. Toxics 2022, 10, 13. https://0-doi-org.brum.beds.ac.uk/10.3390/toxics10010013

AMA Style

Durham T, Guo J, Cowell W, Riley KW, Wang S, Tang D, Perera F, Herbstman JB. Prenatal PM2.5 Exposure in Relation to Maternal and Newborn Telomere Length at Delivery. Toxics. 2022; 10(1):13. https://0-doi-org.brum.beds.ac.uk/10.3390/toxics10010013

Chicago/Turabian Style

Durham, Teresa, Jia Guo, Whitney Cowell, Kylie W. Riley, Shuang Wang, Deliang Tang, Frederica Perera, and Julie B. Herbstman. 2022. "Prenatal PM2.5 Exposure in Relation to Maternal and Newborn Telomere Length at Delivery" Toxics 10, no. 1: 13. https://0-doi-org.brum.beds.ac.uk/10.3390/toxics10010013

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