Next Article in Journal
In-Treatment Changes in Quality of Life-Related Variables in Therapeutic Communities for Cocaine Abusers: Are These Changes Associated with Clinical Outcomes?
Previous Article in Journal
Effectiveness of Swimming Program in Adolescents with Down Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Environmental Air Pollutants Inhaled during Pregnancy Are Associated with Altered Cord Blood Immune Cell Profiles

by
Gabriela Martins Costa Gomes
1,
Wilfried Karmaus
2,
Vanessa E. Murphy
1,
Peter G. Gibson
3,4,
Elizabeth Percival
1,
Philip M. Hansbro
3,5,
Malcolm R. Starkey
6,
Joerg Mattes
1,7 and
Adam M. Collison
1,*
1
Priority Research Centre GrowUpWell®, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2308, Australia
2
School of Public Health, University of Memphis, Memphis, TN 38152, USA
3
Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW 2308, Australia
4
Sleep Medicine Department, John Hunter Hospital, Newcastle, NSW 2305, Australia
5
Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, NSW 2007, Australia
6
Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
7
Paediatric Respiratory & Sleep Medicine Department, John Hunter Children’s Hospital, Newcastle, NSW 2305, Australia
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2021, 18(14), 7431; https://doi.org/10.3390/ijerph18147431
Submission received: 24 May 2021 / Revised: 2 July 2021 / Accepted: 7 July 2021 / Published: 12 July 2021
(This article belongs to the Section Children's Health)

Abstract

:
Air pollution exposure during pregnancy may be a risk factor for altered immune maturation in the offspring. We investigated the association between ambient air pollutants during pregnancy and cell populations in cord blood from babies born to mothers with asthma enrolled in the Breathing for Life Trial. For each patient (n = 91), daily mean ambient air pollutant levels were extracted during their entire pregnancy for sulfur dioxide (SO2), nitric oxide, nitrogen dioxide, carbon monoxide, ozone, particulate matter <10 μm (PM10) or <2.5 μm (PM2.5), humidity, and temperature. Ninety-one cord blood samples were collected, stained, and assessed using fluorescence-activated cell sorting (FACS). Principal Component (PC) analyses of both air pollutants and cell types with linear regression were employed to define associations. Considering risk factors and correlations between PCs, only one PC from air pollutants and two from cell types were statistically significant. PCs from air pollutants were characterized by higher PM2.5 and lower SO2 levels. PCs from cell types were characterized by high numbers of CD8 T cells, low numbers of CD4 T cells, and by high numbers of plasmacytoid dendritic cells (pDC) and low numbers of myeloid DCs (mDCs). PM2.5 levels during pregnancy were significantly associated with high numbers of pDCs (p = 0.006), and SO2 with high numbers of CD8 T cells (p = 0.002) and low numbers of CD4 T cells (p = 0.011) and mDCs (p = 4.43 × 10−6) in cord blood. These data suggest that ambient SO2 and PM2.5 exposure are associated with shifts in cord blood cell types that are known to play significant roles in inflammatory respiratory disease in childhood.

1. Introduction

Asthma is the most common medical condition during pregnancy, with up to 45% of pregnant women with asthma requiring medical care for it [1,2]. Maternal asthma has been found to increase the risk of adverse neonatal outcomes, including respiratory complications [3,4,5], and it has been reported that prenatal exposure to air pollutants such as particulate matter, ozone, and nitrogen oxides increased the risk of transient tachypnea of the newborn, asphyxia, and respiratory distress syndrome [6].
Air pollution refers to the mixture of gases and particulate matter (PM) composed of organic chemicals, metals, gases, biological agents, volatile organic compounds, and minerals carried in the air [7,8,9], which collectively have been linked to harm in nearly every organ in the body [10,11]. Around 91% of the world’s population lives in areas where the levels of air pollutants exceed the World Health Organization (WHO) safe limits [12]. The primary gaseous contaminants are carbon monoxide (CO) and dioxide (CO2), nitrogen dioxide (NO2), ozone (O3), and sulfur dioxide (SO2) [13]. According to the WHO, it is estimated that environmental exposure to PM is responsible for significant morbidity and mortality, including ~16% of lung cancer and 11% of chronic obstructive lung disease (COPD) deaths, and more than 20% of ischemic heart disease and stroke [12].
There is a growing body of epidemiological evidence that links exposure to outdoor air pollution and worsening of pre-existing asthma. These changes have been linked to enhanced inflammation through both innate and adaptive immune pathways as reviewed by Bontinck et al. [14]. Such immunological shifts will alter the interaction between the maternal and developing infant immune systems, and in line with the developmental origins of disease hypothesis, have the potential to shape the future susceptibility to disease [15]. The placenta is a natural barrier between mother and fetus during pregnancy; however, it is not an impenetrable barrier, and environmental air pollutants can cross the placenta; therefore, recent studies have explored the impact of air pollutant exposure during gestation on the unborn child [16,17]. Environmental air pollutants particles that translocate into and cross the placental barrier may also indirectly impact the developing immune system of the child through altering the maternal immune environment [16,17]. Maternal exposure to deleterious environmental factors may negatively impact the developing fetus either directly or indirectly and has been shown to have specific effect on birth weight [18,19,20], potentially altering immune cell maturation and function, subsequently influencing the risk of postnatally acquiring inflammatory and allergic diseases [21].
Neonatal susceptibility to environmental pollutants may be due to either direct or indirect effects on various cell types that exert influence over key processes, including cell differentiation, proliferation, and/or maturation [22]. Previous studies have demonstrated that environmental factors including NO2, SO2, PM < 10 μm in diameter (PM10), PM < 2.5 μm (PM2.5), and polycyclic aromatic hydrocarbons (PAHs) are associated with different cell populations measured in cord blood [23,24,25,26,27]. For instance, global cord blood lymphocyte levels and activity are associated with prenatal air pollution exposure to PAHs and PM2.5 [23,28,29]. Thus, early exposure to air pollution and environmental contaminants may affect the newborn immune system [30]. As many of these changes persist throughout life, alterations in the development of the immune system may be linked to an increased risk of an allergic phenotype in childhood and beyond [31,32,33].
Although there are reports of the effects of environmental factors on cord blood cell populations, previous studies did not phenotype the cell populations in great detail [23,24,25,26,27,28,29,34] and did not account for the influence that maternal asthma has on the newborn immune system [35]. Thus, we investigated the association between levels of ambient air pollutants during pregnancy (SO2, nitric oxide (NO), NO2, CO, O3, PM10, and PM2.5) on populations of well-defined cord blood cells using exposure dimensionality-reduction methods (principal component analysis, PCA) and linear regression approaches to assess associations between maternal exposure and cord blood cell populations from babies born to asthmatic mothers.

2. Materials and Methods

2.1. Study Design and Participants

Pregnant asthmatic women, 18 years or older, with physician-diagnosed asthma, were enrolled in the Breathing for Life Trial (BLT) [36]. The BLT is a multicenter (Brisbane (QLD), Canberra (ACT), Newcastle (NSW), and Sydney (NSW)) randomized controlled trial of asthma management during pregnancy, with follow-up into childhood. Maternal drug or alcohol dependence, chronic oral corticosteroid use, chronic lung disease other than asthma, concomitant chronic illness were exclusion criteria. Eligible mothers agreed to have an interviewer-administered questionnaire conducted during enrolment and information ascertained covered sociodemographic characteristics and lifestyle factors. Participants self-reported age, ethnicity, parity, health status, drug/alcohol dependence. Height and weight were also measured during the first visit. Enrolled mothers at the Newcastle site who consented to participate in the infant follow-up had cord blood collected after delivery. Trained staff extracted information from medical records on gestational age at birth, birth weight, birth length, mode of delivery, maternal and neonatal complications. In this study, 91 pregnant asthmatic woman and their babies were included from mothers previously enrolled in the Breathing for Life Trial (Figure S1).

2.2. Ethics Statement

The study was approved by the Hunter New England Human Research Ethics Committee (Ref no 12/10/17/3.04) and all women provided written informed consent before participation.

2.3. Cord Blood Collection

Cord blood samples were collected at John Hunter Hospital (New Lambton Heights, NSW, Australia) immediately after birth (n = 91) by needle puncture of the umbilical vein after the umbilical cord was detached from the infant. All samples were transferred into EDTA tubes to be processed by trained staff within six hours.

2.4. Flow Cytometry Analysis

Cord blood cells were stained in whole blood and subsets were pre-defined based on specific surface markers as follows: Eosinophils (CD45+, CD193+, CD16), neutrophils (CD45+, CD193, CD16+), CD4 T lymphocytes (CD3+, αβ T-cell receptor [TCR]+, CD4+), CD8 T lymphocytes (CD3+, αβTCR+, CD8+), regulatory T (Treg) cells (CD3+, αβTCR+, CD4+, CD25+, CD127), natural killer (NK) cells (CD14, CD3, CD56+, CD16+), myeloid dendritic cells (mDCs—CD3, CD19, CD56, CD14, HLA-DR+, CD303, CD16+/−, CD1c+/−, CD141+), plasmacytoid dendritic cells (pDCs—CD3, CD19, CD56, CD14, HLA-DR+, CD303+), innate lymphoid cells (ILCs) type 1 (ILC1—CD45+, lineage-negative (Lin; CD3, TCR-αβ, TCR-γδ, CD19, CD11c, CD94, CD14, CD1a, CD34, CD123, CD303, FcεRIα), CD127+, CD161+, CD117, CRTh2, NKp44), ILCs type 2 (ILC2—CD45+, Lin, CD127+, CD161+, CRTh2high, CD117), ILCs type 3 (ILC3) natural-cytotoxicity-receptor-negative (NCR; CD45+, Lin, CD127+, CD161+, CD294, CD117+, NKp44), ILC3 natural-cytotoxicity-receptor-positive (NCR+; CD45+, Lin, CD127+, CD161+, CD294, CD117+, NKp44+) (Table S1). After 30 min of incubation, red blood cells were lysed, and cells were fixed using BD FACS™ Lysing Solution and washed. Samples were stored at 4 °C and acquired within 48 h on a LSRFortessa X-20 flow cytometer (BD Biosciences, San Diego, CA, USA). For the granulocyte panel, NK, lymphocytes and DCs panels a total of 1,000,000 events were acquired and recorded for each subject. The ILCs panels had a total of 2,500,000 events recorded for each subject. Analyses of cell types were conducted with FlowJo software (v 10.5, Flow Jo LLC, Ashland, OR, USA). Results are shown as positive cells in 103 of CD45 positive cells (for granulocytes, ILCs), as positive cells in 103 of CD3 positive cells (NK cells, lymphocytes), and as positive cells in 103 of HLA-DR (DCs).

2.5. Air Pollutant Assessment

Maternal exposure to air pollutants throughout pregnancy was approximated using data from the New South Wales (NSW) Air Quality Monitoring Network [37]. NSW air quality monitoring is achieved through an extensive network of National Association of Testing Authorities-accredited air quality monitoring stations. It reports the data as ambient concentrations and air quality index values, which are stored in a searchable public database. The data undergo rigorous quality assurance processes to ensure reliability. Quality assurance procedures are implemented, both in-the-field and post-data-collection, to ensure that air quality and meteorological parameters measured by the Office of Environment and Heritage air quality monitoring network are reliable. Data are available for the duration of the study period 2017–2019. All air pollution monitors used within this study contribute data on a regular basis and data are updated every morning [37,38].
Air pollutant levels were extracted as the mean daily level across pregnancy from hourly measurements. Levels were obtained for SO2, NO, NO2, CO, O3, PM10, PM2.5, humidity, and temperature. For each air pollutant, the trimesters’ mean level throughout pregnancy was normalized as a quotient of the hourly temperature and humidity at the time of collection prior to further analysis.
Air pollution exposure data during pregnancy and the distribution of prenatal risk factors are shown in Table 1. The relationship among the pollutants is shown in Figure 1.

2.6. Statistical Analysis

Analyses were performed using Stata IC 16.1 (Stata Corporation, College Station, TX, USA). Prior to the analysis of individual air pollutants and cord blood cells, to avoid multicollinearity, PCs of the seven air pollutants were identified. A similar approach was conducted to capture latent features or correlated cells. Through this multivariate analysis, it was possible to select subgroups of pollutants and cells that comprised multiple correlated individual markers and explain their variance [39]. The PCA approach first linearly re-arranged the original correlated variables into fewer new integrated variables (PCs). After varimax rotation, the PCA reduced the data dimensionality and increased interpretability and minimized information loss creating new uncorrelated variables that successively maximized the variance of the PC capturing the seven air pollutants. Hence, PCA found the most informative or explanatory features hidden in the data without a priori knowledge and reduced the number of tests needed for multiple pollutants.
PCA analysis was first applied for air pollutants (SO2, NO, NO2, CO, O3, PM10, PM2.5). This approach was also applied to cell types (eosinophils, neutrophils, Treg cells, CD4 T cells, CD8 T cells, NK cell, pDCs, mDCs, ILC1, CRTh2high ILC2, NCR ILC3, NCR+ ILC3), focusing on factors with an eigenvalue of ≥1. Linear regression analyses were then applied to identify significant associations between the PCs of air pollutants and cell types. Here, PCs were considered for further analysis when p < 0.05. PCs previously selected were subsequently adjusted by potential confounders (other risk factors).
From the PCs considered for further analysis, individual components were selected based on PC loadings to further understand the specific air pollutants and cell types’ relationships. Here, PCs with a cut-off of ±0.4 were considered for further multivariable regression models. The following covariates of interest, based on work by Lura and collaborators [27], were included in the multiregression model: (a) male sex, (b) gestational age, (c) maternal smoking during pregnancy, (d) parity, (e) fetal heart rate decelerations during labor, and (f) mode of delivery. To adjust for multiple comparisons with Bonferroni correction, the error rate (0.05) was divided by the number of tests [40], which varied between the two multivariable regressions applied with the final cut off for significance in each analysis identified in the table legend.

3. Results

3.1. PCA from Air Pollutants Have Positive Loading for O3 and PM2.5

To condense the information of a large number of variables into a smaller set of new composite dimensions, with a minimum loss of information, PCAs were applied to air pollutants and cell types separately. Two PCs were retained on the PCA for the analysis of inhaled air pollutants during pregnancy. These two PCs account for 75.0% of the variance in the original seven variables. The first PC is described by having positive loading for O3 and the second had higher loadings for PM2.5 (Table 2, Figure S2).

3.2. PCA from Cell Types Have Positive Loading for CD8 T cells, ILC1, CRTh2high ILC2, NCR ILC3, Neutrophils, pDCs, and NCR+ ILC3

For the analysis of cord blood cell types, four PCs were retained in the PCA, which together accounted for 60.7% of the variation in the original 12 variables. PC1 presented high numbers of CD8 T cells, while PC2 had higher ILC1, CRTh2high ILC2, and NCR ILC3 cell numbers. The third PC3 was loaded with neutrophils, and the fourth was characterized by pDCs and NCR+ ILC3 (Table 3, Figures S3 and S4).

3.3. Air Pollutant PC2 Associates with Cell Type PC1 and Cell Type PC4

To avoid multiple testing between all air pollutants and all cell types, the analysis first focused on associations between PCs of air pollutants and PCs of cell types. To this end, we conducted eight tests, not adjusting for potential confounders. These crude associations were considered for further regression analysis, if their p-value was <0.05 divided by the number of tests (p < 0.006); this resulted in three significant associations (Table 4).
After further adjustment for prenatal risk factors (male sex, gestational age, maternal smoking during pregnancy, parity, fetal heart rate decelerations during labor, and mode of delivery) in regression models, only associations between air pollutant PC2 and cell type PC1 (β = −0.429, CI = −0.618 to −0.239, p = 2.19 × 10−5); and air pollutant PC2 and cell type PC4 (β = 0.471, CI = 0.273 to 0.668, p = 8.91 × 10−6) maintained statistical significance (Table 5). Thus, only these two associations were considered for further analysis.

3.4. SO2 Associates with CD8 T Cells, CD4 T Cells and mDCs While PM2.5 Associates with pDCs in a Multipollutant Multivariable Regression Models

After screening for potential associations using PCs capturing air pollutants and a distinct PC capturing cell counts, the next steps focused on individual air pollutants and cell types identified in the associations between the PCs. The only PC from air pollutants associated with cell type PCs in the multivariable regression model was PC2 (Table 5). Thus, only PC2 from air pollutants was considered for the multipollutant multi-cell analyses (Table 2 and Table 5).
Regarding the two significant PC of the cells, CD4 T cells and CD8 T cells were considered for further analysis from PC1. For PC4 of the cells, pDC and NCR+ ILC3 had loadings >0.4, and mDCs presented loadings < −0.4 (Table 3 and Table 5, Figures S3 and S4). For this model, the following potential confounders were taken into consideration: male sex, gestational age, maternal smoking during pregnancy, parity, fetal heart rate decelerations during labor, and mode of delivery, with the addition of the concentration of the pollutants with the magnitude of 0.4 (PM2.5 and SO2).
In the second multipollutant multivariable regression model that considered pollutants selected in PC2 (air pollutants) and cells selected in PC1 (cell types), SO2 positively correlated with CD8 T cells (β = 260.242, CI = 99.087 to 421.397, p = 0.002), and negatively associated with CD4 T cells (β = −234.283, CI = −413.582 to −54.984, p = 0.011; Table 6). Here, applying the Bonferroni correction to four tests the critical p-value was 0.012.
Analyzing individual components of PC2 (air pollutants) and of PC4 (cell types), a positive association was found between PM2.5 and pDCs (β = 155.158, CI = 46.578 to 263.738, p = 0.006) and between SO2 and mDCs (β = 879.250, CI = 523.834 to 1234.666, p = 4.43 × 10−06). For this analysis with six tests, after Bonferroni correction, statistical significance was considered when p was less than 0.008 (Table 7).

4. Discussion

During pregnancy, the fetus has an intense and prolonged interaction with the mother at the maternal–fetus interface. Within this period, a complex network of interactions which provide passive immunity to the newborn, program the neonatal immune system, and tune its homeostatic regulation, is formed [35,41]. Maternal asthma represents a unique risk factor for childhood health, and asthmatic individuals are more susceptible to environmental exposures which can both interact with the maternal immune system and cross the placenta to directly interact with the developing child with detrimental health effects previously reported from early life onwards [16,17]. In this study, of the air pollutants investigated, only SO2 and PM2.5 were associated with differences in any of the 12 pre-defined cord blood cell populations after screening employing dimensionality-reduction. Mean daily local SO2 levels during gestation were negatively associated with CD4 T cells in cord blood, and positively associated with both CD8 T cells and mDCs numbers. In addition, mean daily local PM2.5 levels through pregnancy were positively associated with pDCs in the cord blood.
T-lymphocytes play an important role in the immune system, tailoring the body’s immune response to specific pathogens through the release of regulatory cytokines. T-cell development starts during the early weeks of gestation [32] during which toxic exposure can result in failure of stem cell formation or, in the later phases of gestation, can cause abnormal stem cell formation, interrupting cell migration and proliferation [42,43,44]. Based on cytokine production, activated CD4 T cells are predominantly classified as either T helper (Th) type 1 (Th1) lymphocytes that produce interferon (IFN)-γ, interleukin (IL)-2 and IL-12, or Th type 2 (Th2) lymphocytes that produce mainly IL-4, IL-5, and IL-13 [45]. The counter-regulation between Th1 and Th2 is capable of inhibiting or inducing the development of an allergic phenotype [46,47]. Exposure to immunotoxic compounds in utero can cause immuno-suppression and predispose to aberrant immune responses later in life [48].
Gaseous molecules can freely diffuse through biological membranes and SO2 inhalation can cause oxidative injury in the cardiovascular, respiratory, digestive, reproductive, endocrine, and neurological systems [49]. Several studies point to the relationship between SO2 exposure and inflammation; SO2 levels were significantly increased in acute pneumonia and chronic renal failure patients [50,51], and pro-inflammatory cytokines levels were increased in the lungs of mice after exposure to SO2 [52].
In this study, SO2 negatively associated with cord blood CD4 T cells, while positively associating with CD8 T cells. Air pollutant exposure during the early months of pregnancy has previously been shown to influence the Th1/Th2 homeostatic balance [23], and levels of cell proliferation in the cord blood have also been reported to be associated with air pollutant exposure ex vivo [53]. A recent experimental study showed epithelial damage and increased infiltration of inflammatory cells into the airways after PM2.5 exposure and also an immune imbalance of Th cells [54]. The exposure to PM2.5 also disturbs the balance of T helper 17 (Th17)/Treg cells [55]. Both acute and long-term exposure to high levels of PM2.5 were associated with alterations in differentially methylated regions of forkhead box P3 (Foxp3) [56].
Previous studies conducted in Australia report with similar levels of PM2.5 and identify an association between air pollutant exposure and pregnancy disorders such as hypertensive disorders during pregnancy and increased likelihood of gestational diabetes mellitus [57] which can further affect the fetus [58,59]. Another study conducted in the United States, also showing similar levels of PM2.5 in similar cohorts, has demonstrated that later phases of prenatal lung development may be particularly sensitive to the developmental toxicity of PM2.5 [60]. Although air pollution is a universal issue, it is likely that there are high-risk individuals who are susceptible to the greatest harm when exposed to PM2.5 [61]. It has further shown that PM2.5 can induce allergic airway inflammation [62], and trigger exacerbations in pre-existing asthma and COPD [63]. As smaller particles, PM2.5 is readily able to penetrate deeper into the lungs and cross into the bloodstream at a higher rate than larger particles [64], which can cause chronic inflammation in pregnant women and hamper fetal development [21,65,66]. Air pollution exposure during pregnancy has been linked to placental inflammation and impairment of placental function [67]. PM2.5 exposure may cause inflammatory responses in the placenta, which may be transmitted from the mother to the fetus and contribute to the development of abnormalities [68,69].
Interestingly, DC subsets quantified in the cord blood positively associated with environmental SO2 as well as PM2.5. DCs are a major link between the innate and the adaptive immune system. They recognize antigens through the expression of innate receptors such as toll-like receptor (TLR), and process and present fragments of these antigens on their cell surface to T-lymphocytes that then deliver effector responses. Studies show that a pro-inflammatory response in the airway mediated by TLR activation, might be stimulated by PM-associated biological components, such as pollen, bacteria, fungal spores, and viruses, as well as with soluble metals, and organic content [70,71,72,73,74]. Here, air pollutant measurements were taken in a region of Australia and included months with potentially increased air pollution due to environmental events such as bushfires which may also have contributed to the alterations seen in DC subsets [75,76,77,78].
PM acts on APCs, such as DCs, as an adjuvant. Cultured DCs stimulated by PM increase maturation with elevated expression of CD80 and major histocompatibility complex class II (MHC-II) and increased pro-inflammatory cytokine release [79,80]. PM-stimulation also promotes DC expression of C-C chemokine receptor type 7 (CCR7), which directs lymph-node homing [81] initiating the immune response cascade. Thus, enhanced DC maturation may promote an enhanced T-lymphocyte response to PM [79,82,83]. In individuals with existing asthma, increased pDC numbers and activity have been linked to acute exacerbations, particularly virally induced exacerbations [84]. Indeed, pDC function may play a significant role in the pathogenesis of asthma. We used animal models to demonstrate that pDCs deficient in TLR7 contribute to virally induced exacerbations of allergic airways disease that is reversed by TLR7 competent pDCs [85]. Thus, it is tempting to speculate that the increased pDC levels found in children whose mothers had highest exposure to PM2.5 may contribute to an increased susceptibility to develop virally induced wheeze and asthma later in life. This will be of interest as our cohort grows to an age where these measures can be evaluated. Children born to mothers with moderate to severe uncontrolled asthma during pregnancy are at increased risk of developing asthma and more commonly have lung function abnormalities [4,5]. However, studies have shown that with better asthma control during pregnancy and fewer exacerbations, there was a better respiratory health outcome for the children [86,87], suggesting that these pathways may be modified through improved maternal asthma management.
During pregnancy, maternal immune responses shift towards a type 2 (T2) predominance that promotes immunological tolerance towards the fetus. The balance of type 1 (T1) and T2 cytokines in pregnancy is thought to be crucial to maternal tolerance of the infant [88,89]. The fetal immune system is thought to be under the direct influence of the maternal immune response mounted at the fetus–maternal interface and studies have demonstrated that asthmatic individuals are susceptible to environmental exposure, and air pollution can cause exacerbations of pre-existing asthma [90,91,92]. Air pollution exposure during pregnancy was previously shown to be associated with reduced postnatal lung function [93], and it has been demonstrated that the effect of prenatal air pollution exposure on lung function at five weeks are sustained up to 11 years of age [94]. Perinatal air pollution exposure was also demonstrated to affect asthma onset during pre-school and school age periods [60,95]. It is emerging that there might be further life-long implications with several recent studies having shown that lung function in early life tracks into adulthood [96,97,98,99], and is associated with an increased risk of chronic respiratory diseases including asthma and COPD [100].
The findings observed in this study are limited to pregnancies in mothers with asthma, and the air pollution data available were generalized from local air pollution monitoring stations which are limited in their accuracy to the actual levels in the air breathed by the participants in their homes and workplaces. This study shares the limited sample size that is a common limitation of cord blood studies. Our sample included cord blood of 91 newborns, however, the advantages are the ability to access a suitable quantity of blood so early in life. This has enabled cord blood studies to make significant contributions to our understanding of early life immune and respiratory development [101,102,103,104,105,106]. In addition, all samples collected and analyzed in this study were from a population at high-risk of developing lung disease, who were infants born to asthmatic mothers, which may provide increased power to detect immune changes associated with subsequent lung disease.
In summary, higher levels of inhaled SO2 during pregnancy may have a direct effect, reducing cord blood CD4 T cells while increasing CD8 T cells and mDCs. It is suggested that inhaled PM2.5 exert their effects by increasing cord blood pDCs numbers. Through the evaluation of the association between ambient air pollutants during pregnancy and cord blood immune cell types, this study shows that SO2 and PM2.5 exposure during pregnancy are associated with shifts in cord blood cell types which may cause an inflammatory response in the placenta that may influence fetal development. Further follow-up studies of this cohort and complimentary mechanistic studies are required to elucidate if these immunological shifts persist into later life and/or are associated with increased risk of subsequent chronic disease.

5. Conclusions

In summary, higher levels of inhaled SO2 during pregnancy are associated with reduced cord blood CD4 T cells and increased CD8 T cells and mDCs. Local PM2.5 levels through pregnancy were also associated with increased cord blood pDCs numbers. Through the evaluation of the association between ambient air pollutants during pregnancies complicated by asthma and cord blood immune cell types, this study shows that SO2 and PM2.5 exposure is associated with shifts in cord blood cell types that are known to play significant roles in inflammatory respiratory disease in childhood. Further follow-up studies of this cohort and complimentary mechanistic studies are required to elucidate if these immunological shifts persist into later life and/or are associated with subsequent chronic disease.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijerph18147431/s1, Table S1: Antibodies used in flow cytometry analysis, Figure S1: Flow chart. Recruitment, collection of cord blood samples and maternal air pollutant exposure assessment. FACS, fluorescence-activated cell sorting, Figure S2: Component loadings (A) and score variables (B) from the PCA (air pollutant) analyses, Figure S3: Component loadings from the PCA (cell type) analyses, Figure S4: Score variables from the PCA (cell type) analyses.

Author Contributions

P.G.G., V.E.M., J.M. and A.M.C. conducted BLT study; A.M.C. conceived the project and supervised analyses; W.K. reviewed statistical analyses; G.M.C.G., A.M.C., W.K., E.P., M.R.S., P.M.H. and J.M. worked on study design. G.M.C.G. was involved in data collection, drafted the manuscript, processed cord blood samples and all analyses involved. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Rebecca L Cooper Research Foundation, the Priority Research Centre GrowUpWell, the National Health and Medical Research Council (NHMRC, 1060983), the Hunter Medical Research Institute and the Hunter Children’s Research Foundation. AMC received a fellowship from The Thoracic Society of Australia and New Zealand and the National Asthma Council. PMH is funded by the NHMRC and Medical Research Future Fund (1175134, 1201338). MRS is funded by the NHMRC (1156898, 20000483) and Australian Research Council (DE170100226).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by Hunter New England Human Research Ethics Committee (Reference Number 12/10/17/3.04, NSW HREC Reference No: HREC/12/ HNE/357).

Informed Consent Statement

Written informed consent has been obtained from the patient(s).

Data Availability Statement

The data presented in this study is available upon request to the corresponding author.

Acknowledgments

We would like to thank the midwives involved at John Hunter Hospital for their collaboration in this project. We thank the whole BLT team. We appreciate comments and technical assistance from A. Pereira de Siqueira, J. Grehan, Nrs. K. Steel, M. Jensen.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murphy, V.E.; Namazy, J.A.; Powell, H.; Schatz, M.; Chambers, C.; Attia, J.; Gibson, P.G. A meta-analysis of adverse perinatal outcomes in women with asthma. BJOG 2011, 118, 1314–1323. [Google Scholar] [CrossRef] [PubMed]
  2. Murphy, V.E. Managing asthma in pregnancy. Breathe 2015, 11, 258–267. [Google Scholar] [CrossRef]
  3. Mendola, P.; Mannisto, T.I.; Leishear, K.; Reddy, U.M.; Chen, Z.; Laughon, S.K. Neonatal health of infants born to mothers with asthma. J. Allergy Clin. Immunol. 2014, 133, 85–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Martel, M.J.; Rey, E.; Beauchesne, M.F.; Malo, J.L.; Perreault, S.; Forget, A.; Blais, L. Control and severity of asthma during pregnancy are associated with asthma incidence in offspring: Two-stage case-control study. Eur. Respir. J. 2009, 34, 579–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Valadares, M.A.; Gurgel, R.Q.; Melo, E.V.; Guimaraes, A.M.; Guedes, K.M.; Rocha, N.A.; Almeida, M.L. Respiratory function in children of asthmatic mothers. J. Pediatr. 2013, 89, 158–163. [Google Scholar] [CrossRef] [Green Version]
  6. Seeni, I.; Ha, S.; Nobles, C.; Liu, D.; Sherman, S.; Mendola, P. Air pollution exposure during pregnancy: Maternal asthma and neonatal respiratory outcomes. Ann. Epidemiol. 2018, 28, 612–618. [Google Scholar] [CrossRef] [PubMed]
  7. Valavanidis, A.; Fiotakis, K.; Vlachogianni, T. Airborne particulate matter and human health: Toxicological assessment and importance of size and composition of particles for oxidative damage and carcinogenic mechanisms. J. Environ. Sci. Health Part C Environ. Carcinog. Ecotoxicol. Rev. 2008, 26, 339–362. [Google Scholar] [CrossRef] [PubMed]
  8. Nemmar, A.; Holme, J.A.; Rosas, I.; Schwarze, P.E.; Alfaro-Moreno, E. Recent advances in particulate matter and nanoparticle toxicology: A review of the in vivo and in vitro studies. Biomed. Res. Int. 2013, 2013, 279371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Idowu, O.; Semple, K.T.; Ramadass, K.; O’Connor, W.; Hansbro, P.; Thavamani, P. Beyond the obvious: Environmental health implications of polar polycyclic aromatic hydrocarbons. Environ. Int. 2019, 123, 543–557. [Google Scholar] [CrossRef]
  10. Schraufnagel, D.E.; Balmes, J.R.; Cowl, C.T.; De Matteis, S.; Jung, S.H.; Mortimer, K.; Perez-Padilla, R.; Rice, M.B.; Riojas-Rodriguez, H.; Sood, A.; et al. Pollution and noncommunicable diseases: A review by the forum of international respiratory societies’ environmental committee, part 1: The damaging effects of air pollution. Chest 2019, 155, 409–416. [Google Scholar] [CrossRef] [Green Version]
  11. Schraufnagel, D.E.; Balmes, J.R.; Cowl, C.T.; De Matteis, S.; Jung, S.H.; Mortimer, K.; Perez-Padilla, R.; Rice, M.B.; Riojas-Rodriguez, H.; Sood, A.; et al. 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, 417–426. [Google Scholar] [CrossRef] [PubMed]
  12. Balakrishnan, K.; Dey, S.; Gupta, T.; Dhaliwal, R.S.; Brauer, M.; Cohen, A.J.; Stanaway, J.D.; Beig, G.; Joshi, T.K.; Aggarwal, A.N.; et al. The impact of air pollution on deaths, disease burden, and life expectancy across the states of India: The Global Burden of Disease Study 2017. Lancet Planet. Health 2019, 3, e26–e39. [Google Scholar] [CrossRef] [Green Version]
  13. Morakinyo, O.M.; Mokgobu, M.I.; Mukhola, M.S.; Hunter, R.P. Health outcomes of exposure to biological and chemical components of inhalable and respirable particulate matter. Int. J. Environ. Res. Public Health 2016, 13, 592. [Google Scholar] [CrossRef] [PubMed]
  14. Bontinck, A.; Maes, T.; Joos, G. Asthma and air pollution: Recent insights in pathogenesis and clinical implications. Curr. Opin. Pulm. Med. 2020, 26, 10–19. [Google Scholar] [CrossRef]
  15. Heindel, J.J.; Vandenberg, L.N. Developmental origins of health and disease: A paradigm for understanding disease cause and prevention. Curr. Opin. Pediatr. 2015, 27, 248. [Google Scholar] [CrossRef] [Green Version]
  16. Koren, G.; Ornoy, A. The role of the placenta in drug transport and fetal drug exposure. Expert Rev. Clin. Pharm. 2018, 11, 373–385. [Google Scholar] [CrossRef]
  17. Bove, 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]
  18. Darrow, L.A.; Klein, M.; Strickland, M.J.; Mulholland, J.A.; Tolbert, P.E. Ambient air pollution and birth weight in full-term infants in Atlanta, 1994–2004. Environ. Health Perspect 2011, 119, 731–737. [Google Scholar] [CrossRef] [Green Version]
  19. Maisonet, M.; Bush, T.J.; Correa, A.; Jaakkola, J.J. Relation between ambient air pollution and low birth weight in the Northeastern United States. Environ. Health Perspect 2001, 109 (Suppl. 3), 351–356. [Google Scholar] [CrossRef] [Green Version]
  20. Trasande, L.; Wong, K.; Roy, A.; Savitz, D.A.; Thurston, G. Exploring prenatal outdoor air pollution, birth outcomes and neonatal health care utilization in a nationally representative sample. J. Expo. Sci. Environ. Epidemiol. 2013, 23, 315–321. [Google Scholar] [CrossRef] [Green Version]
  21. Glinianaia, S.V.; Rankin, J.; Bell, R.; Pless-Mulloli, T.; Howel, D. Particulate air pollution and fetal health: A systematic review of the epidemiologic evidence. Epidemiology 2004, 15, 36–45. [Google Scholar] [CrossRef]
  22. Pinkerton, K.E.; Joad, J.P. Influence of air pollution on respiratory health during perinatal development. Clin. Exp. Pharm. Physiol. 2006, 33, 269–272. [Google Scholar] [CrossRef] [PubMed]
  23. Herr, C.E.; Dostal, M.; Ghosh, R.; Ashwood, P.; Lipsett, M.; Pinkerton, K.E.; Sram, R.; Hertz-Picciotto, I. Air pollution exposure during critical time periods in gestation and alterations in cord blood lymphocyte distribution: A cohort of livebirths. Environ. Health 2010, 9, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hertz-Picciotto, I.; Dostal, M.; Dejmek, J.; Selevan, S.G.; Wegienka, G.; Gomez-Caminero, A.; Sram, R.J. Air pollution and distributions of lymphocyte immunophenotypes in cord and maternal blood at delivery. Epidemiology 2002, 13, 172–183. [Google Scholar] [CrossRef] [PubMed]
  25. Hertz-Picciotto, I.; Herr, C.E.; Yap, P.S.; Dostal, M.; Shumway, R.H.; Ashwood, P.; Lipsett, M.; Joad, J.P.; Pinkerton, K.E.; Sram, R.J. Air pollution and lymphocyte phenotype proportions in cord blood. Environ. Health Perspect. 2005, 113, 1391–1398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Baiz, N.; Slama, R.; Bene, M.C.; Charles, M.A.; Kolopp-Sarda, M.N.; Magnan, A.; Thiebaugeorges, O.; Faure, G.; Annesi-Maesano, I. Maternal exposure to air pollution before and during pregnancy related to changes in newborn’s cord blood lymphocyte subpopulations. The EDEN study cohort. BMC Pregnancy Childbirth 2011, 11, 87. [Google Scholar] [CrossRef] [Green Version]
  27. Lura, M.P.; Gorlanova, O.; Muller, L.; Proietti, E.; Vienneau, D.; Reppucci, D.; Pavlovic, R.; Dahinden, C.; Roosli, M.; Latzin, P.; et al. Response of cord blood cells to environmental, hereditary and perinatal factors: A prospective birth cohort study. PLoS ONE 2018, 13, e0200236. [Google Scholar] [CrossRef]
  28. Herr, C.E.; Ghosh, R.; Dostal, M.; Skokanova, V.; Ashwood, P.; Lipsett, M.; Joad, J.P.; Pinkerton, K.E.; Yap, P.S.; Frost, J.D.; et al. Exposure to air pollution in critical prenatal time windows and IgE levels in newborns. Pediatr. Allergy Immunol. 2011, 22, 75–84. [Google Scholar] [CrossRef]
  29. Leonardi, G.S.; Houthuijs, D.; Steerenberg, P.A.; Fletcher, T.; Armstrong, B.; Antova, T.; Lochman, I.; Lochmanova, A.; Rudnai, P.; Erdei, E.; et al. Immune biomarkers in relation to exposure to particulate matter: A cross-sectional survey in 17 cities of Central Europe. Inhal. Toxicol. 2000, 12 (Suppl. 4), 1–14. [Google Scholar] [CrossRef]
  30. Ashley-Martin, J.; Lavigne, E.; Arbuckle, T.E.; Johnson, M.; Hystad, P.; Crouse, D.L.; Marshall, J.S.; Dodds, L. Air pollution during pregnancy and cord blood immune system biomarkers. J. Occup. Environ. Med. 2016, 58, 979–986. [Google Scholar] [CrossRef] [Green Version]
  31. Dietert, R.R.; Lee, J.E.; Hussain, I.; Piepenbrink, M. Developmental immunotoxicology of lead. Toxicol. Appl. Pharm. 2004, 198, 86–94. [Google Scholar] [CrossRef]
  32. Holladay, S.D.; Smialowicz, R.J. Development of the murine and human immune system: Differential effects of immunotoxicants depend on time of exposure. Environ. Health Perspect. 2000, 108, 463–473. [Google Scholar] [CrossRef] [Green Version]
  33. Luebke, R.W.; Chen, D.H.; Dietert, R.; Yang, Y.; King, M.; Luster, M.I.; Immunotoxicology, W. The comparative immunotoxicity of five selected compounds following developmental or adult exposure. J. Toxicol. Environ. Health 2006, 9, 1–26. [Google Scholar] [CrossRef]
  34. Latzin, P.; Frey, U.; Armann, J.; Kieninger, E.; Fuchs, O.; Roosli, M.; Schaub, B. Exposure to moderate air pollution during late pregnancy and cord blood cytokine secretion in healthy neonates. PLoS ONE 2011, 6, e23130. [Google Scholar] [CrossRef] [Green Version]
  35. Jennewein, M.F.; Abu-Raya, B.; Jiang, Y.; Alter, G.; Marchant, A. Transfer of maternal immunity and programming of the newborn immune system. Semin. Immunopathol. 2017, 39, 605–613. [Google Scholar] [CrossRef]
  36. Murphy, V.E.; Jensen, M.E.; Mattes, J.; Hensley, M.J.; Giles, W.B.; Peek, M.J.; Bisits, A.; Callaway, L.K.; McCaffery, K.; Barrett, H.L.; et al. The Breathing for Life Trial: A randomised controlled trial of fractional exhaled nitric oxide (FENO)-based management of asthma during pregnancy and its impact on perinatal outcomes and infant and childhood respiratory health. BMC Pregnancy Childbirth 2016, 16, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. NSW Department of Planning, Industry and Environment. New South Wales Air Quality Monitoring Network. Available online: https://www.environment.nsw.gov.au (accessed on 8 April 2020).
  38. Riley, M.; Kirkwood, J.; Jiang, N.; Ross, G.; Scorgie, Y. Air quality monitoring in NSW: From long term trend monitoring to integrated urban services. Air Qual. Clim. Chang. 2020, 54, 44–51. [Google Scholar]
  39. Ramsay, J.O.; Silverman, B.W. Principal components analysis for functional data. In Functional Data Analysis; Springer: New York, NY, USA, 2005; pp. 147–172. [Google Scholar]
  40. McDonald, J.H. Handbook of Biological Statistics, 3rd ed.; Sparky House Publishing: Baltimore, MA, USA, 2014. [Google Scholar]
  41. Ghazal, P.; Dickinson, P.; Smith, C.L. Early life response to infection. Curr. Opin. Infect. Dis. 2013, 26, 213–218. [Google Scholar] [CrossRef]
  42. Bakker, J.M.; Schmidt, E.D.; Kroes, H.; Kavelaars, A.; Heijnen, C.J.; Tilders, F.J.; Van Rees, E.P. Effects of short-term dexamethasone treatment during pregnancy on the development of the immune system and the hypothalamo-pituitary adrenal axis in the rat. J. Neuroimmunol. 1995, 63, 183–191. [Google Scholar] [CrossRef]
  43. Landreth, K.S. Critical windows in development of the rodent immune system. Hum. Exp. Toxicol. 2002, 21, 493–498. [Google Scholar] [CrossRef] [PubMed]
  44. Vanders, R.L.; Murphy, V.E.; Gibson, P.G.; Hansbro, P.M.; Wark, P.A. CD8 T cells and dendritic cells: Key players in the attenuated maternal immune response to influenza infection. J. Reprod. Immunol. 2015, 107, 1–9. [Google Scholar] [CrossRef]
  45. Kaiko, G.E.; Horvat, J.C.; Beagley, K.W.; Hansbro, P.M. Immunological decision-making: How does the immune system decide to mount a helper T-cell response? Immunology 2008, 123, 326–338. [Google Scholar] [CrossRef] [PubMed]
  46. Maggi, E.; Parronchi, P.; Manetti, R.; Simonelli, C.; Piccinni, M.P.; Rugiu, F.S.; De Carli, M.; Ricci, M.; Romagnani, S. Reciprocal regulatory effects of IFN-gamma and IL-4 on the in vitro development of human Th1 and Th2 clones. J. Immunol. 1992, 148, 2142–2147. [Google Scholar] [PubMed]
  47. Thorburn, A.N.; Hansbro, P.M. Harnessing regulatory T cells to suppress asthma: From potential to therapy. Am. J. Respir. Cell Mol. Biol. 2010, 43, 511–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Dietert, R.R.; Etzel, R.A.; Chen, D.; Halonen, M.; Holladay, S.D.; Jarabek, A.M.; Landreth, K.; Peden, D.B.; Pinkerton, K.; Smialowicz, R.J.; et al. Workshop to identify critical windows of exposure for children’s health: Immune and respiratory systems work group summary. Environ. Health Perspect. 2000, 108, 483–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Wang, X.B.; Du, J.B.; Cui, H. Sulfur dioxide, a double-faced molecule in mammals. Life Sci. 2014, 98, 63–67. [Google Scholar] [CrossRef] [PubMed]
  50. Kajiyama, H.; Nojima, Y.; Mitsuhashi, H.; Ueki, K.; Tamura, S.; Sekihara, T.; Wakamatsu, R.; Yano, S.; Naruse, T. Elevated levels of serum sulfite in patients with chronic renal failure. J. Am. Soc. Nephrol. 2000, 11, 923–927. [Google Scholar] [CrossRef]
  51. Mitsuhashi, H.; Ikeuchi, H.; Yamashita, S.; Kuroiwa, T.; Kaneko, Y.; Hiromura, K.; Ueki, K.; Nojima, Y. Increased levels of serum sulfite in patients with acute pneumonia. Shock 2004, 21, 99–102. [Google Scholar] [CrossRef]
  52. Meng, Z.; Liu, Y.; Wu, D. Effect of sulfur dioxide inhalation on cytokine levels in lungs and serum of mice. Inhal. Toxicol. 2005, 17, 303–307. [Google Scholar] [CrossRef] [PubMed]
  53. Novack, L.; Yitshak-Sade, M.; Landau, D.; Kloog, I.; Sarov, B.; Karakis, I. Association between ambient air pollution and proliferation of umbilical cord blood cells. Environ. Res. 2016, 151, 783–788. [Google Scholar] [CrossRef] [PubMed]
  54. Shen, Y.; Zhang, Z.H.; Hu, D.; Ke, X.; Gu, Z.; Zou, Q.Y.; Hu, G.H.; Song, S.H.; Kang, H.Y.; Hong, S.L. The airway inflammation induced by nasal inoculation of PM2.5 and the treatment of bacterial lysates in rats. Sci. Rep. 2018, 8, 9816. [Google Scholar] [CrossRef] [PubMed]
  55. Sun, L.; Fu, J.; Lin, S.H.; Sun, J.L.; Xia, L.; Lin, C.H.; Liu, L.; Zhang, C.; Yang, L.; Xue, P.; et al. Particulate matter of 2.5 mum or less in diameter disturbs the balance of TH17/regulatory T cells by targeting glutamate oxaloacetate transaminase 1 and hypoxia-inducible factor 1alpha in an asthma model. J. Allergy Clin. Immunol. 2020, 145, 402–414. [Google Scholar] [CrossRef] [Green Version]
  56. Prunicki, M.; Stell, L.; Dinakarpandian, D.; de Planell-Saguer, M.; Lucas, R.W.; Hammond, S.K.; Balmes, J.R.; Zhou, X.; Paglino, T.; Sabatti, C.; et al. Exposure to NO2, CO, and PM2.5 is linked to regional DNA methylation differences in asthma. Clin. Epigenetics 2018, 10, 2. [Google Scholar] [CrossRef] [Green Version]
  57. Melody, S.M.; Wills, K.; Knibbs, L.D.; Ford, J.; Venn, A.; Johnston, F. Maternal Exposure to Ambient Air Pollution and Pregnancy Complications in Victoria, Australia. Int. J. Environ. Res. Public Health 2020, 17, 2572. [Google Scholar] [CrossRef] [Green Version]
  58. Cunningham, M.W., Jr.; LaMarca, B. Risk of cardiovascular disease, end-stage renal disease, and stroke in postpartum women and their fetuses after a hypertensive pregnancy. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2018, 315, R521–R528. [Google Scholar] [CrossRef] [PubMed]
  59. Ornoy, A.; Reece, E.A.; Pavlinkova, G.; Kappen, C.; Miller, R.K. Effect of maternal diabetes on the embryo, fetus, and children: Congenital anomalies, genetic and epigenetic changes and developmental outcomes. Birth Defects Res. Embryo Today 2015, 105, 53–72. [Google Scholar] [CrossRef] [PubMed]
  60. Hazlehurst, M.F.; Carroll, K.N.; Loftus, C.T.; Szpiro, A.A.; Moore, P.E.; Kaufman, J.D.; Kirwa, K.; LeWinn, K.Z.; Bush, N.R.; Sathyanarayana, S.; et al. Maternal exposure to PM2.5 during pregnancy and asthma risk in early childhood: Consideration of phases of fetal lung development. Environ. Epidemiol. 2021, 5, e130. [Google Scholar] [CrossRef]
  61. Tong, S. Air pollution and disease burden. Lancet Planet. Health 2019, 3, e49–e50. [Google Scholar] [CrossRef] [Green Version]
  62. Ogino, K.; Nagaoka, K.; Ito, T.; Takemoto, K.; Okuda, T.; Nakayama, S.F.; Ogino, N.; Seki, Y.; Hamada, H.; Takashiba, S.; et al. Involvement of PM2.5-bound protein and metals in PM2.5-induced allergic airway inflammation in mice. Inhal. Toxicol. 2018, 30, 498–508. [Google Scholar] [CrossRef]
  63. Falcon-Rodriguez, C.I.; Osornio-Vargas, A.R.; Sada-Ovalle, I.; Segura-Medina, P. Aeroparticles, composition, and lung diseases. Front. Immunol. 2016, 7, 3. [Google Scholar] [CrossRef] [Green Version]
  64. Nemmar, A.; Hoet, P.H.; Vanquickenborne, B.; Dinsdale, D.; Thomeer, M.; Hoylaerts, M.F.; Vanbilloen, H.; Mortelmans, L.; Nemery, B. Passage of inhaled particles into the blood circulation in humans. Circulation 2002, 105, 411–414. [Google Scholar] [CrossRef] [Green Version]
  65. Rogers, J.F.; Thompson, S.J.; Addy, C.L.; McKeown, R.E.; Cowen, D.J.; Decoufle, P. Association of very low birth weight with exposures to environmental sulfur dioxide and total suspended particulates. Am. J. Epidemiol. 2000, 151, 602–613. [Google Scholar] [CrossRef] [PubMed]
  66. Sram, R.J.; Binkova, B.; Dejmek, J.; Bobak, M. Ambient air pollution and pregnancy outcomes: A review of the literature. Environ. Health Perspect. 2005, 113, 375–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Dexter, S.C.; Pinar, H.; Malee, M.P.; Hogan, J.; Carpenter, M.W.; Vohr, B.R. Outcome of very low birth weight infants with histopathologic chorioamnionitis. Obs. Gynecol. 2000, 96, 172–177. [Google Scholar] [CrossRef]
  68. Yue, H.; Ji, X.; Li, G.; Hu, M.; Sang, N. Maternal exposure to PM2.5 affects fetal lung development at sensitive windows. Environ. Sci. Technol. 2020, 54, 316–324. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, Y.; Wang, L.; Wang, F.; Li, C. Effect of fine particulate matter (PM2.5) on rat placenta pathology and perinatal outcomes. Med. Sci. Monit. 2016, 22, 3274–3280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Shoenfelt, J.; Mitkus, R.J.; Zeisler, R.; Spatz, R.O.; Powell, J.; Fenton, M.J.; Squibb, K.A.; Medvedev, A.E. Involvement of TLR2 and TLR4 in inflammatory immune responses induced by fine and coarse ambient air particulate matter. J. Leukoc. Biol. 2009, 86, 303–312. [Google Scholar] [CrossRef] [Green Version]
  71. Becker, S.; Fenton, M.J.; Soukup, J.M. Involvement of microbial components and toll-like receptors 2 and 4 in cytokine responses to air pollution particles. Am. J. Respir. Cell Mol. Biol. 2002, 27, 611–618. [Google Scholar] [CrossRef]
  72. Goulaouic, S.; Foucaud, L.; Bennasroune, A.; Laval-Gilly, P.; Falla, J. Effect of polycyclic aromatic hydrocarbons and carbon black particles on pro-inflammatory cytokine secretion: Impact of PAH coating onto particles. J. Immunotoxicol. 2008, 5, 337–345. [Google Scholar] [CrossRef]
  73. Hopke, P.K.; Rossner, A. Exposure to airborne particulate matter in the ambient, indoor, and occupational environments. Clin. Occup. Environ. Med. 2006, 5, 747–771. [Google Scholar] [CrossRef]
  74. Inoue, K.; Takano, H.; Yanagisawa, R.; Hirano, S.; Ichinose, T.; Shimada, A.; Yoshikawa, T. The role of toll-like receptor 4 in airway inflammation induced by diesel exhaust particles. Arch. Toxicol. 2006, 80, 275–279. [Google Scholar] [CrossRef]
  75. Yu, P.; Xu, R.; Abramson, M.J.; Li, S.; Guo, Y. Bushfires in Australia: A serious health emergency under climate change. Lancet Planet. Health 2020, 4, e7–e8. [Google Scholar] [CrossRef] [Green Version]
  76. Nogrady, B. Bushfires: Australia issues health warnings as Sydney air quality plummets. BMJ 2019, 367, l6914. [Google Scholar] [CrossRef] [PubMed]
  77. Vardoulakis, S.; Marks, G.; Abramson, M.J. Lessons learned from the australian bushfires: Climate change, air pollution, and public health. JAMA Intern. Med. 2020, 180, 635–636. [Google Scholar] [CrossRef] [PubMed]
  78. Dharwal, V.; Paudel, K.R.; Hansbro, P.M. Impact of bushfire smoke on respiratory health. Med. J. Aust 2020, 213, 284. [Google Scholar] [CrossRef]
  79. Porter, M.; Karp, M.; Killedar, S.; Bauer, S.M.; Guo, J.; Williams, D.; Breysse, P.; Georas, S.N.; Williams, M.A. Diesel-enriched particulate matter functionally activates human dendritic cells. Am. J. Respir. Cell Mol. Biol. 2007, 37, 706–719. [Google Scholar] [CrossRef] [Green Version]
  80. Matthews, N.C.; Faith, A.; Pfeffer, P.; Lu, H.; Kelly, F.J.; Hawrylowicz, C.M.; Lee, T.H. Urban particulate matter suppresses priming of T helper type 1 cells by granulocyte/macrophage colony-stimulating factor-activated human dendritic cells. Am. J. Respir. Cell Mol. Biol. 2014, 50, 281–291. [Google Scholar] [CrossRef]
  81. Pfeffer, P.E.; Ho, T.R.; Mann, E.H.; Kelly, F.J.; Sehlstedt, M.; Pourazar, J.; Dove, R.E.; Sandstrom, T.; Mudway, I.S.; Hawrylowicz, C.M. Urban particulate matter stimulation of human dendritic cells enhances priming of naive CD8 T lymphocytes. Immunology 2018, 153, 502–512. [Google Scholar] [CrossRef]
  82. Bleck, B.; Tse, D.B.; Jaspers, I.; Curotto de Lafaille, M.A.; Reibman, J. Diesel exhaust particle-exposed human bronchial epithelial cells induce dendritic cell maturation. J. Immunol. 2006, 176, 7431–7437. [Google Scholar] [CrossRef] [Green Version]
  83. Bleck, B.; Tse, D.B.; Curotto de Lafaille, M.A.; Zhang, F.; Reibman, J. Diesel exhaust particle-exposed human bronchial epithelial cells induce dendritic cell maturation and polarization via thymic stromal lymphopoietin. J. Clin. Immunol. 2008, 28, 147–156. [Google Scholar] [CrossRef]
  84. Chairakaki, A.D.; Saridaki, M.I.; Pyrillou, K.; Mouratis, M.A.; Koltsida, O.; Walton, R.P.; Bartlett, N.W.; Stavropoulos, A.; Boon, L.; Rovina, N.; et al. Plasmacytoid dendritic cells drive acute asthma exacerbations. J. Allergy Clin. Immunol. 2018, 142, 542–556. [Google Scholar] [CrossRef] [Green Version]
  85. Hatchwell, L.; Collison, A.; Girkin, J.; Parsons, K.; Li, J.; Zhang, J.; Phipps, S.; Knight, D.; Bartlett, N.W.; Johnston, S.L.; et al. Toll-like receptor 7 governs interferon and inflammatory responses to rhinovirus and is suppressed by IL-5-induced lung eosinophilia. Thorax 2015, 70, 854–861. [Google Scholar] [CrossRef] [Green Version]
  86. Morten, M.; Collison, A.; Murphy, V.E.; Barker, D.; Oldmeadow, C.; Attia, J.; Meredith, J.; Powell, H.; Robinson, P.D.; Sly, P.D.; et al. Managing Asthma in Pregnancy (MAP) trial: FeNO levels and childhood asthma. J. Allergy Clin. Immunol. 2018, 142, 1765–1772. [Google Scholar] [CrossRef] [Green Version]
  87. Mattes, J.; Murphy, V.E.; Powell, H.; Gibson, P.G. Prenatal origins of bronchiolitis: Protective effect of optimised asthma management during pregnancy. Thorax 2014, 69, 383–384. [Google Scholar] [CrossRef] [Green Version]
  88. Halonen, M.; Lohman, I.C.; Stern, D.A.; Spangenberg, A.; Anderson, D.; Mobley, S.; Ciano, K.; Peck, M.; Wright, A.L. Th1/Th2 patterns and balance in cytokine production in the parents and infants of a large birth cohort. J. Immunol. 2009, 182, 3285–3293. [Google Scholar] [CrossRef] [PubMed]
  89. Lehmann, I.; Herberth, G. Cord blood immune status: Predicting health or allergy? Allergy 2012, 67, 445–448. [Google Scholar] [CrossRef] [PubMed]
  90. O’Connor, G.T.; Neas, L.; Vaughn, B.; Kattan, M.; Mitchell, H.; Crain, E.F.; Evans, R., 3rd; Gruchalla, R.; Morgan, W.; Stout, J.; et al. Acute respiratory health effects of air pollution on children with asthma in US inner cities. J. Allergy Clin. Immunol. 2008, 121, 1133–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Weinmayr, G.; Romeo, E.; De Sario, M.; Weiland, S.K.; Forastiere, F. Short-term effects of PM10 and NO2 on respiratory health among children with asthma or asthma-like symptoms: A systematic review and meta-analysis. Environ. Health Perspect. 2010, 118, 449–457. [Google Scholar] [CrossRef] [Green Version]
  92. Tiotiu, A.I.; Novakova, P.; Nedeva, D.; Chong-Neto, H.J.; Novakova, S.; Steiropoulos, P.; Kowal, K. Impact of Air Pollution on Asthma Outcomes. Int. J. Environ. Res. Public Health 2020, 17, 6212. [Google Scholar] [CrossRef] [PubMed]
  93. Latzin, P.; Roosli, M.; Huss, A.; Kuehni, C.E.; Frey, U. Air pollution during pregnancy and lung function in newborns: A birth cohort study. Eur. Respir. J. 2009, 33, 594–603. [Google Scholar] [CrossRef] [Green Version]
  94. Korten, I.; Ramsey, K.; Latzin, P. Air pollution during pregnancy and lung development in the child. Paediatr. Respir. Rev. 2017, 21, 38–46. [Google Scholar] [CrossRef]
  95. Sbihi, H.; Tamburic, L.; Koehoorn, M.; Brauer, M. Perinatal air pollution exposure and development of asthma from birth to age 10 years. Eur. Respir. J. 2016, 47, 1062–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Stern, D.A.; Morgan, W.J.; Wright, A.L.; Guerra, S.; Martinez, F.D. Poor airway function in early infancy and lung function by age 22 years: A non-selective longitudinal cohort study. Lancet 2007, 370, 758–764. [Google Scholar] [CrossRef] [Green Version]
  97. Turner, S.W.; Palmer, L.J.; Rye, P.J.; Gibson, N.A.; Judge, P.K.; Cox, M.; Young, S.; Goldblatt, J.; Landau, L.I.; Le Souef, P.N. The relationship between infant airway function, childhood airway responsiveness, and asthma. Am. J. Respir. Crit. Care Med. 2004, 169, 921–927. [Google Scholar] [CrossRef]
  98. Filippone, M.; Sartor, M.; Zacchello, F.; Baraldi, E. Flow limitation in infants with bronchopulmonary dysplasia and respiratory function at school age. Lancet 2003, 361, 753–754. [Google Scholar] [CrossRef]
  99. Hoo, A.F.; Stocks, J.; Lum, S.; Wade, A.M.; Castle, R.A.; Costeloe, K.L.; Dezateux, C. Development of lung function in early life: Influence of birth weight in infants of nonsmokers. Am. J. Respir. Crit. Care Med. 2004, 170, 527–533. [Google Scholar] [CrossRef] [PubMed]
  100. Martinez, F.D. The origins of asthma and chronic obstructive pulmonary disease in early life. Proc. Am. Thorac. Soc. 2009, 6, 272–277. [Google Scholar] [CrossRef] [Green Version]
  101. Junge, K.M.; Hornig, F.; Herberth, G.; Roder, S.; Kohajda, T.; Rolle-Kampczyk, U.; von Bergen, M.; Borte, M.; Simon, J.C.; Heroux, D.; et al. The LINA cohort: Cord blood eosinophil/basophil progenitors predict respiratory outcomes in early infancy. Clin. Immunol. 2014, 152, 68–76. [Google Scholar] [CrossRef] [PubMed]
  102. Gern, J.E.; Calatroni, A.; Jaffee, K.F.; Lynn, H.; Dresen, A.; Cruikshank, W.W.; Lederman, H.M.; Sampson, H.A.; Shreffler, W.; Bacharier, L.B.; et al. Patterns of immune development in urban preschoolers with recurrent wheeze and/or atopy. J. Allergy Clin. Immunol. 2017, 140, 836–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hornsby, E.; Pfeffer, P.E.; Laranjo, N.; Cruikshank, W.; Tuzova, M.; Litonjua, A.A.; Weiss, S.T.; Carey, V.J.; O’Connor, G.; Hawrylowicz, C. Vitamin D supplementation during pregnancy: Effect on the neonatal immune system in a randomized controlled trial. J. Allergy Clin. Immunol. 2018, 141, 269–278. [Google Scholar] [CrossRef] [Green Version]
  104. Bullens, D.M.; Seys, S.; Kasran, A.; Dilissen, E.; Dupont, L.J.; Ceuppens, J.L. Low cord blood Foxp3/CD3gamma mRNA ratios: A marker of increased risk for allergy development. Clin. Exp. Allergy 2015, 45, 232–237. [Google Scholar] [CrossRef] [PubMed]
  105. Perveen, K.; Quach, A.; McPhee, A.; Prescott, S.L.; Barry, S.C.; Hii, C.S.; Ferrante, A. Validation of monoclonal anti-PKC isozyme antibodies for flow cytometry analyses in human T cell subsets and expression in cord blood T cells. Sci. Rep. 2019, 9, 9263. [Google Scholar] [CrossRef] [PubMed]
  106. Ludwig, E.; Harder, J.; Lacorcia, M.; Honkpehedji, Y.J.; Nouatin, O.P.; van Dam, G.J.; Corstjens, P.; Sartono, E.; Esen, M.; Lobmaier, S.M.; et al. Placental gene expression and antibody levels of mother-neonate pairs reveal an enhanced risk for inflammation in a helminth endemic country. Sci. Rep. 2019, 9, 15776. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Correlation plots between SO2, NO, NO2, CO, O3, PM10, and PM2.5. Air pollutant levels are represented as the mean daily level across pregnancy from hourly measurements. Each air pollutant was normalized as a quotient of the hourly temperature and humidity at the time of collection.
Figure 1. Correlation plots between SO2, NO, NO2, CO, O3, PM10, and PM2.5. Air pollutant levels are represented as the mean daily level across pregnancy from hourly measurements. Each air pollutant was normalized as a quotient of the hourly temperature and humidity at the time of collection.
Ijerph 18 07431 g001
Table 1. Population characteristics.
Table 1. Population characteristics.
Mean
(Min–Max)/n (%)
Total
(n)
Demographic Characteristics
Maternal smoking during pregnancy11 (12.1)91
Maternal recurrent asthma exacerbation during pregnancy4 (4.9)81
Maternal age at delivery30.0 (19.0–41.5)91
Gestational age at delivery (weeks)39 (34–41)91
Caesarean section33 (36.3)90
Mode of labor, spontaneous16 (17.8)90
Mode of labor, augmented9 (10.0)90
Mode of labor, induced48 (53.3)90
Fetal heart rate decelerations during labor29 (32.2)90
Male sex49 (53.8)91
Older siblings46 (51.1)90
Birth weight (kg)3.5 (2.1–4.9)90
Birth length (cm)51.6 (30.7–58.0)86
Air pollution exposure during pregnancy—Mean daily concentration
SO2 (µg/m³)4.2 (3.5–5.0)91
PM10 (µg/m³)23.6 (20.4–26.8)91
PM2.5 (µg/m³)7.8 (7.1–8.5)91
NO (µg/m³)7.9 (4.1–9.7)91
NO2 (µg/m³)15.0 (10.2–17.6)91
CO (µg/m³)0.3 (0.2–0.4)91
O3 (µg/m³)39.0 (34.6–44.4)91
Humidity (%)69.9 (66.1–73.9)91
Temperature (°C)18.6 (16.8–20.7)91
SO2 sulfur dioxide, PM10 particulate matter < 10 µm in diameter, PM2.5 particulate matter < 2.5 µm in diameter, NO nitric oxide, NO2 nitrogen dioxide, CO carbon monoxide, O3 ozone.
Table 2. Components loadings for air pollutant PC1 and PC2 after varimax rotation.
Table 2. Components loadings for air pollutant PC1 and PC2 after varimax rotation.
Air PollutantsPC1PC2
SO20.144−0.719
NO−0.464−0.002
NO2−0.4300.260
CO0.2680.051
O30.505−0.197
PM100.3960.386
PM2.50.3090.474
SO2 sulfur dioxide, PM10 particulate matter < 10 µm in diameter, PM2.5 particulate matter < 2.5 µm in diameter, NO nitric oxide, NO2 nitrogen dioxide, CO carbon monoxide, O3 ozone.
Table 3. Component loadings for cell types PC1, PC2, PC3, and PC4 after varimax rotation.
Table 3. Component loadings for cell types PC1, PC2, PC3, and PC4 after varimax rotation.
Cell TypesPC1PC2PC3PC4
Eosinophils0.0060.045−0.683−0.005
Neutrophils−0.0020.0510.698−0.014
Treg−0.325−0.1250.0360.304
TCD4−0.583−0.018−0.018−0.163
TCD80.5570.058−0.055−0.003
NK cells0.362−0.2340.110−0.050
pDC0.183−0.064−0.0630.585
mDC0.1960.0470.071−0.402
ILC1−0.1720.4880.0410.064
CRTh2high ILC20.0630.564−0.093−0.158
NCR ILC30.0860.5860.0780.115
NCR+ ILC30.0120.1230.0650.577
Table 4. Crude regression model of PCs of air pollutants and cord blood cell types. After Bonferroni correction, significance was considered when p < 0.006.
Table 4. Crude regression model of PCs of air pollutants and cord blood cell types. After Bonferroni correction, significance was considered when p < 0.006.
Crude
Regression
PC1
Cell Types
PC2
Cell Types
PC3
Cell Types
PC4
Cell Types
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p ValueCoef.
(95% CI)
p
Value
PC1
Air pollutant
−0.021
(−0.173; 0.129)
0.7760.018
(−0.129; 0.165)
0.804−0.055
(−0.198; 0.087)
0.4430.083
(−0.053; 0.219)
0.230
PC2
Air pollutant
−0.411
(−0.632; −0.190)
0.0004−0.187
(−0.415; 0.040)
0.106−0.320
(−0.535; −0.106)
0.0040.495
(0.305; 0.684)
1.29 × 10−6
Table 5. Multivariable regression model of PCs of air pollutants previously associated with PCs of cord blood cell types in an univariable regression model. After Bonferroni correction, significance was considered when p < 0.016.
Table 5. Multivariable regression model of PCs of air pollutants previously associated with PCs of cord blood cell types in an univariable regression model. After Bonferroni correction, significance was considered when p < 0.016.
Multivariable Regression *PC1
Cell Types
PC3
Cell Types
PC4
Cell Types
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p
Value
PC2
Air pollutant
−0.429
(−0.618; −0.239)
2.19 × 10−5−0.192
(−0.390; 0.005)
0.0560.471
(0.273; 0.668)
8.91 × 10−6
Coef. coefficient; CI confidence interval. * Adjusted for male sex, gestational age, maternal smoking during pregnancy, parity, fetal heart rate deceleration during labor, mode of delivery.
Table 6. Multipollutant multivariable regression models of pollutants and cord blood cells previously associated in PCA. PC2 from air pollutants and PC1 from cell types with ≥0.4 for the highest loadings and ≤−0.4 for the lowest loadings. After Bonferroni correction, significance was considered when p < 0.012.
Table 6. Multipollutant multivariable regression models of pollutants and cord blood cells previously associated in PCA. PC2 from air pollutants and PC1 from cell types with ≥0.4 for the highest loadings and ≤−0.4 for the lowest loadings. After Bonferroni correction, significance was considered when p < 0.012.
Multipollutant Multivariable Model *
PM2.5SO2
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p
Value
TCD8 −4.268
(−104.138; 95.602)
0.932260.242
(99.087; 421.397)
0.002
TCD4 63.128
(−47.986; 174.242)
0.262−234.283
(−413.582–54.984)
0.011
PM2.5 particulate matter < 2.5 µm in diameter; SO2 sulfur dioxide; Coef coefficient; CI confidence interval; * Adjusted for male sex, gestational age, maternal smoking during pregnancy, parity, fetal heart rate deceleration during labor, mode of delivery, SO2, and PM2.5. Results are expressed in 103 of CD3 positive cells.
Table 7. Multipollutant multivariable regression models of cord blood cells and pollutants previously associated in PCA. PC2 from air pollutants and PC4 from cell types with ≥0.4 for the highest loadings and ≤−0.4 for the lowest loadings. After Bonferroni correction, significance was considered when p < 0.008.
Table 7. Multipollutant multivariable regression models of cord blood cells and pollutants previously associated in PCA. PC2 from air pollutants and PC4 from cell types with ≥0.4 for the highest loadings and ≤−0.4 for the lowest loadings. After Bonferroni correction, significance was considered when p < 0.008.
Multipollutant Multivariable Model *
PM2.5SO2
Coef.
(95% CI)
p
Value
Coef.
(95% CI)
p
Value
pDC 155.158
(46.578; 263.738)
0.006−169.378
(−344.588; 5.832)
0.058
mDC −90.800
(−311.056, 129.456)
0.414879.250
(523.834; 1234.666)
4.43 × 10−6
NCR + ILC3 0.149
(0.006; 0.290)
0.0410.011
(−0.219; 0.240)
0.927
PM2.5 particulate matter < 2.5 µm in diameter; PM10 particulate matter < 10 µm in diameter; SO2 sulfur dioxide; Coef coefficient; CI confidence interval; * Adjusted for male sex, gestational age, maternal smoking during pregnancy, parity, fetal heart rate deceleration during labor, mode of delivery, SO2, and PM2.5. Results are expressed in 103 of HLA-DR positive cells. Results are expressed in 103 of CD45 positive cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martins Costa Gomes, G.; Karmaus, W.; Murphy, V.E.; Gibson, P.G.; Percival, E.; Hansbro, P.M.; Starkey, M.R.; Mattes, J.; Collison, A.M. Environmental Air Pollutants Inhaled during Pregnancy Are Associated with Altered Cord Blood Immune Cell Profiles. Int. J. Environ. Res. Public Health 2021, 18, 7431. https://doi.org/10.3390/ijerph18147431

AMA Style

Martins Costa Gomes G, Karmaus W, Murphy VE, Gibson PG, Percival E, Hansbro PM, Starkey MR, Mattes J, Collison AM. Environmental Air Pollutants Inhaled during Pregnancy Are Associated with Altered Cord Blood Immune Cell Profiles. International Journal of Environmental Research and Public Health. 2021; 18(14):7431. https://doi.org/10.3390/ijerph18147431

Chicago/Turabian Style

Martins Costa Gomes, Gabriela, Wilfried Karmaus, Vanessa E. Murphy, Peter G. Gibson, Elizabeth Percival, Philip M. Hansbro, Malcolm R. Starkey, Joerg Mattes, and Adam M. Collison. 2021. "Environmental Air Pollutants Inhaled during Pregnancy Are Associated with Altered Cord Blood Immune Cell Profiles" International Journal of Environmental Research and Public Health 18, no. 14: 7431. https://doi.org/10.3390/ijerph18147431

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