Next Article in Journal
VAV1 Gene Polymorphisms in Patients with Rheumatoid Arthritis
Previous Article in Journal
Ambient Air Pollution Exposure Association with Anaemia Prevalence and Haemoglobin Levels in Chinese Older Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Influence of Second-Hand Smoke and Prenatal Tobacco Smoke Exposure on Biomarkers, Genetics and Physiological Processes in Children—An Overview in Research Insights of the Last Few Years

Institute of Occupational Medicine, Social Medicine and Environmental Medicine, Goethe University Frankfurt, D-60590 Frankfurt, Germany
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2020, 17(9), 3212; https://doi.org/10.3390/ijerph17093212
Submission received: 26 March 2020 / Revised: 16 April 2020 / Accepted: 29 April 2020 / Published: 5 May 2020
(This article belongs to the Section Children's Health)

Abstract

:
Children are commonly exposed to second-hand smoke (SHS) in the domestic environment or inside vehicles of smokers. Unfortunately, prenatal tobacco smoke (PTS) exposure is still common, too. SHS is hazardous to the health of smokers and non-smokers, but especially to that of children. SHS and PTS increase the risk for children to develop cancers and can trigger or worsen asthma and allergies, modulate the immune status, and is harmful to lung, heart and blood vessels. Smoking during pregnancy can cause pregnancy complications and poor birth outcomes as well as changes in the development of the foetus. Lately, some of the molecular and genetic mechanisms that cause adverse health effects in children have been identified. In this review, some of the current insights are discussed. In this regard, it has been found in children that SHS and PTS exposure is associated with changes in levels of enzymes, hormones, and expression of genes, micro RNAs, and proteins. PTS and SHS exposure are major elicitors of mechanisms of oxidative stress. Genetic predisposition can compound the health effects of PTS and SHS exposure. Epigenetic effects might influence in utero gene expression and disease susceptibility. Hence, the limitation of domestic and public exposure to SHS as well as PTS exposure has to be in the focus of policymakers and the public in order to save the health of children at an early age. Global substantial smoke-free policies, health communication campaigns, and behavioural interventions are useful and should be mandatory.

1. Introduction

Second-hand smoke (SHS) consists of mainstream smoke exhaled by a smoker and side-stream smoke from the smouldering tobacco product [1]. The terms “environmental tobacco smoke (ETS)” and “passive smoke” as synonyms for SHS are common but were also described as the sum of SHS and third-hand smoke [2]. Important sources of SHS exposure are the workplace, public places where smoking is allowed, smoker’s homes and vehicles. In particular, homes and vehicles are loci where children and pregnant women can be exposed to SHS [3]. Unfortunately, exposure to SHS in private homes and vehicles is still common [4,5].
SHS is linked to lots of health hazards. More than 7000 chemicals, including at least 70 carcinogenic substances, have been identified in SHS [3]. These carcinogens can cause several types of cancer (e.g., lung cancer, breast cancer, stomach cancer, cancers of the upper respiratory tract) [6,7]. Therefore, SHS increases the risk of children contracting lymphoma, leukaemia, liver cancer or brain tumours [8]. Additionally, SHS is harmful to heart and blood vessels, so it increases the risks of stroke and heart diseases in later life significantly [9]. SHS irritates the airways, can cause or worsen asthmatic diseases and allergies and is a major risk factor in chronic obstructive pulmonary disease (COPD). Tobacco smoke can also trigger lung infections and wheezing in children [10,11]. Exposed very young children are at increased risk of sudden infant death syndrome [8]. Not least, SHS seems to be linked with mental health effects [12]. Prenatal tobacco smoke (PTS) exposure can cause poor birth outcomes and pregnancy complications. PTS is linked with biochemical changes in the placenta, leading to alterations to the antioxidant system of the foetus, associated with several adverse health effects both prenatal and postnatal. PTS exposure can lead to pulmonary diseases, e.g., COPD, wheezing, asthma, kidney diseases as well as cardiovascular diseases in later life. Also, metabolic syndrome and obesity can be a consequence of PTS exposure [8,13].
Extensive data from epidemiological and experimental studies indicate that gene–environmental interaction during pregnancy and early life can induce permanent changes in physiological processes and disease predisposition by epigenetic mechanisms [14]. The early events during pregnancy and childhood play a key role in the development of the human body. In this vulnerable phase, exposure to tobacco smoke have been shown to have deleterious effects on the development process and may result in permanent damage [15]. The genetic predisposition can lead to substantially aggravated risk for diseases of children exposed to SHS [16,17,18,19]. It might be said that PTS is the first major environmental factor that can jeopardise the health of the unborn child (Figure 1) [13].
According to the World Health Organization (WHO), data from 2004 from 192 countries showed that 40% of children and about 33% of adult non-smokers were exposed to SHS worldwide [4,20]. In the United States, the exposition to SHS in non-smokers decreased from 52.5% from 1999 to 2000 to 25.3% from 2011 to 2012 (SHS exposed children aged 3–11 years: 40.6%) [21]. The European human biomonitoring pilot study (September 2011 to January 2012) reported SHS exposure in children aged 6–11 years from Portugal, Poland, and Romania [22]. Regarding the daily SHS exposure at home, the prevalence in Portugal was 15%, Poland 19.3%, and Romania 21.7%. Each year, more than 1.2 million premature deaths are caused by SHS, 65,000 of which are children. Nevertheless, only 22% of the world population is protected by comprehensive national smoke-free laws [23]. Lau and Celermajer [24] declared in 2014, 50 years after the US Surgeon General’s first report in 1964, that the protection of children from SHS is one of the great healthcare challenges.
This review explores a range of basic science research regarding biomarkers, physiological processes (Table 1) and genetics addressing PTS and SHS exposure. Additionally, a short overview will be given on changes in DNA methylation induced by PTS exposure. Molecular and genetic mechanisms underlying tobacco smoke-induced diseases are not completely understood, so further research will be necessary. However, this review aims to inform the policymakers and the public for a deeper understanding of the adverse health effects of tobacco smoke, especially in children.

2. Biomarkers

2.1. Matrix Metalloproteinase-9

The enzyme matrix metalloproteinase-9 (MMP-9), also known as gelatinase B, has the ability to regulate the activity of some soluble proteins and plays a role in the degradation of extracellular matrix components. It plays important parts in physiological and pathological processes [25], e.g., in the pathogenesis of allergy [26]. In this respect, levels of MMP-9 in the sputum of patients with chronic bronchitis or asthma were significantly higher than in a control group [27]. In a study from 2011, De et al. [26] investigated the effects of SHS on levels of MMP-9 in nasal secretions of 39 children aged between 7 and 16 years. The authors found that MMP-9 concentrations and activity were significantly higher in the probed nasal secretions of children exposed to SHS. They concluded that SHS might alter in the nasal mucosa the inflammatory reaction, similar to an allergic response. In a recent study, in children with cystic fibrosis (CF) exposed to SHS, it was found that the MMP-9 gene is overexpressed [28].
Yilmaz et al. [29] came to a different conclusion in an investigation of the influence of SHS exposure on nasal and serum inflammatory markers in 150 wheezing children (91 children exposed to lower levels of SHS, 24 children exposed to higher levels and 35 non-exposed children). Admittedly, the authors reported that SHS aggravated respiratory symptoms in exposed wheezing children significantly, albeit without significant influence on inflammatory markers like nasal MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), glutathione, interleukin-8 and 17 (IL-8, IL-17) and serum surfactant protein-D (SP-D).

2.2. Immune-Regulatory Cytokines

Interleukins (ILs), interferons (IFNs) and tumour necrosis factor-alpha (TNF-α) are immune-regulatory cytokines and therefore biomarkers of inflammatory processes [30]. These cytokines are associated, among others, with asthma, allergy and respiratory illness [10,31,32]. Chahal et al. [33] investigated the effect of PTS on levels of IL-1α, IL-6, IL-8 and IL-1 receptor antagonist in dried blood of new-borns. They ascertained an increase in IL-8 levels among neonates exposed to PTS. Additionally, a decrease in IL-1β, IL-4, IL-5, and IFN-γ was described in healthy 1–6-year-old SHS-exposed children [34]. In an investigation of children and adolescents aged 0–17 years, a positive association between high SHS exposure and IL-1β levels in saliva was found [35]. In saliva samples of five-year-old children exposed to SHS, the levels of IL-1β, IL-6 and TNF-α, but not IL-8, were elevated [36]. Airway IL-13 secretion was increased in SHS-exposed children [37].
The findings of these studies indicate that PTS and SHS exposure influences the levels of immune-regulatory cytokines with effects on inflammatory processes in children.

2.3. Cysteinyl Leukotrienes and Urinary Leukotriene E4

Cysteinyl leukotrienes (CysLTs) are key mediators and modulators in the pathogenesis of asthma [38]. Changes in CysLT expression and excretion can be detected by measuring urinary leukotriene E4 (uLTE4) levels [39]. In smokers, as well as in children exposed to tobacco smoke, leukotriene receptor antagonists (LTRAs) are more effective [40,41]. This shows the CysLT pathway plays an important role in mediating SHS-triggered asthma-related health effects [42]. In 2008, Kott et al. [43] ascertained a correlation between SHS exposure, increase in uLTE4 in exposed infants suffering from respiratory syncytial virus bronchiolitis and length of hospital stay.
In 2011, Rabinovitch et al. [42] proposed uLTE4 as the first biomarker to identify SHS-exposed children at risk of asthma exacerbation. As SHS is linked to increased asthma severity in affected children [44], the biomarker uLTE4 is important to estimate the predisposition of asthmatic children to severe exacerbations. In children with poor asthma control exposed to SHS, a trend for increasing uLTE4 concentrations was determined [45]. A study from 2013 approved the correlation between SHS and increasing uLTE4, but with the indication that the body mass index has still a more pronounced influence on its concentration [46]. One study found that in opposition to inhaled corticosteroids to treat asthma, oral treatment with the LTRA montelukast prevents an increase in uLTE4 level in children at low SHS exposure, but with no effect on asthma control [47]. Meanwhile, it was ascertained that uLTE4 may be useful as a potent biomarker of atopic, viral or IgE-mediated asthma in children [48,49]. It was shown that uLTE4 is also an effective biomarker to determine aspirin intolerance in subjects with asthma [50], and of exposure to atopic and non-atopic asthma triggers, recent exacerbations, and early development of childhood atopy. It is also used for predicting response initiation or step-up therapy with LTRAs [51].

2.4. Estimated Glomerular Filtration Rate and Kidney Function

PTS and SHS exposure are known risk factors for kidney disease [52,53]. In adolescents, Omoloja et al. [54] described SHS exposure not only as a risk factor for chronic kidney disease (CKD) but also as a risk factor independently or together with CKD for adverse cardiovascular outcomes. The estimated glomerular filtration rate (eGFR) as a marker for kidney function was decreased in adolescents exposed to tobacco smoke including SHS and, thus, may affect kidney function in early life [55]. In children with CKD, an association between SHS exposure and nephrotic range proteinuria was found [56]. PTS exposure may lead to lower eGFR and smaller kidney volume in children of school age [57].

2.5. Cardiovascular Status

SHS is a risk factor for coronary heart disease [58]. SHS can lead to endothelial dysfunction, a main factor for cardiovascular disease [59]. Cardiovascular diseases are also caused by PTS exposure [60]. Groner et al. [61] found in 9 to 18 years old children and adolescents that SHS exposure is associated with high levels of soluble intercellular adhesion molecule 1 (s-ICAM1, transmembrane protein released at atherosclerotic lesions) and negatively associated with the prevalence of endothelial progenitor cells. That might lead to both vascular endothelial stress and lower capability to vascular repair. Enlarged carotid artery intima-media thickness (IMT) can be induced by SHS exposure in early life and is a risk factor of atherosclerosis in adulthood [62]. In neonates, it was found that the aortic IMT increased due to PTS exposure [63]. Five-year-old children exposed to PTS had greater carotid IMT than non-exposed [64]. It was found in adults exposed to SHS in childhood that they had greater carotid IMT [65]. In a recent study, it was shown that SHS exposure in childhood leads to an increased risk of atrial fibrillation in adulthood [66].
The described literature shows that exposure to PTS and SHS has negative effects on vascular walls in early life and is harmful to the cardiovascular system also in later life.

2.6. C-Reactive Protein

C-reactive protein (CRP) is involved in the systematic inflammation response. On the other side, CRP is a risk factor for adiposity in children and for the development of atherosclerosis [67,68]. It was shown in non-smoking youths that SHS exposure is associated with a significant increase in CRP serum concentration [69]. Similar results were found in an exploration of 10-year-old children exposed to SHS [70]. High-sensitivity CRP (hsCRP) values in serum were increased in three- to five-year-old SHS-exposed children in a dose-response manner [71]. Additionally, SHS exposure in early life could increase the risk of increased hsCRP levels in adulthood [72].

3. Immune Status

Regulatory T-cells (Tregs) are essentially involved in the immune regulation of atopic diseases [73]. Hinz and colleagues [74] found in a prospective birth cohort study lower Treg cell numbers in cord blood of children exposed to PTS. They concluded this might be a predictor for early atopic dermatitis or sensitization to food allergens in early life. It was shown that maternal smoking during pregnancy leads to a higher expression of microRNA-223 (miR-223) both in maternal blood cells and in cord blood cells. This was, in turn, associated with lower Treg cell numbers [75]. The underlying cause could be the regulative effect of miR-223 on the transcription factor FoxO1 that plays in turn significant roles in T-cell regulation [76,77]. A combination of lower Treg cell numbers and higher numbers of T-helper type 17 (Th17) cells were ascertained in children exposed to SHS [78]. Th17/Treg imbalance is a key component of asthma severity [79]. In SHS-exposed healthy adolescents, a decrease in circulating CD3+ and CD4+ memory cells accompanied by an increase in circulating naïve CD3+ and CD4+ T-cell subsets and absolute CD4+CD45RA+ cell counts in a dose-effect response was found [80]. Whereby, this systemic immunological response could be a possible initiator for diseases in later life. The proportion of IFN-γ producing CD8+ cells was lower in adenoids from SHS-exposed children, possibly resulting in an increasing predisposition to respiratory infections [81]. In an in vitro investigation, it was ascertained that adenoidal B-lymphocytes of SHS exposed and/or atopic children produce more IgA and IgM. For this purpose, B-lymphocytes of ectomised tonsils of children with adenoidal hypertrophy were cultured and stimulated [82].
Yao et al. [83] showed in an investigation of immunoglobulin E (IgE) levels against 40 allergens that SHS exposure of children and adolescents is significantly associated with IgE sensitization to grass pollen, cockroaches, and certain foods (cow’s milk, egg white, crab, shrimp, codfish, soybean, potato, peanut, almond, garlic, and cheese), but not with sensitization against mites, mould, and latex. SHS exposure in early infancy, but not during pregnancy, was associated with an increased risk of food sensitization (IgE antibody reactivity against cow’s milk, egg white, soybean, peanut, cod, and wheat) up to age 16 years [84]. In general, SHS exposure in early childhood increases the risk of allergic sensitization [85].
Kopp et al. [28] showed in infants and young children with cystic fibrosis (CF) disproportionately exposed to SHS that inflammatory gene expression and arachidonic acid (AA) metabolism are altered, resulting in an impaired bacterial clearance. Among others, they also reported an association between SHS exposure and decrease in the AA metabolite prostaglandin D2, suppression of two prostaglandin genes (prostaglandin reductase 2, PTGR2 and prostaglandin E synthase 3, PTGES3), and overexpression of regulatory factor X2 gene.

4. Lipid Profile

It was described that lower serum levels of high-density lipoprotein-cholesterol (HDL-C) in children are associated with SHS exposure, suggesting an increased risk of arteriosclerosis [86]. SHS-exposed children showed significantly higher values of triglycerides, total cholesterol and low-density lipoprotein-cholesterol (LDL-C) in combination with lower values of HDL-C, and enhanced peripheral lymphocyte apoptosis possibly caused by the altered lipid profile [87]. In schoolchildren, an association between SHS exposure, higher values of triglycerides and lower HDL-C levels was reported, meaning an enhanced risk of obesity and metabolic syndrome [88]. HDL-C reductions were also found in SHS-exposed toddlers [89] and in healthy 8-year-old children exposed to PTS [90]. Lower HDL-C levels were also observed in female adolescents, but not in male ones [91]. Another study showed an increase in apolipoprotein (Apo) B and ratio ApoB/ApoA-1 in adolescents exposed to SHS [92]. Accordingly, low ApoA-1 and high lipoprotein-associated phospholipase A2 concentrations were noticed at 10-year-old children exposed to SHS [70]. High triglycerides and low HDL-C levels were found in women 18–44 years after PTS exposure [93]. Regarding lipid metabolism of neonates, several phosphatidylcholine (lipid metabolites) levels were affected by PTS exposure and differed significantly in sera probes from mothers and cord blood [94].
These study results report that PTS and SHS exposure has a negative influence on the lipid profile in children up to adulthood accompanied by associated health effects. In contrast, Zakhar and colleagues [95] reported no material association between SHS exposure and lipid profiles in children. However, the authors concluded that an effect on abnormalities of serum lipid levels might require a longer tobacco smoke exposition time.

5. Oxidative Stress

Oxidative stress (OS) is characterised by increased intracellular levels of reactive oxygen species (ROS) [96]. In severe cases, OS can lead to cell and tissue injury and even cell death [97], and is associated, among others, with asthma [98] and cardiovascular events in adulthood [99]. OS can lead to many foetal and neonate diseases because free radicals, with their harmful effects like cellular, tissue and organ damage, cannot be fought by a weakened antioxidant system [100]. OS plays a key role in the pathogenesis of COPD [11]. SHS is an important inductor of OS, particularly among adolescents and children [101,102].
The enzyme nicotinamide adenine dinucleotide phosphate oxidase-2 (Nox2) forms ROS by generating super-oxide [103]. It was found that Nox2 activity was higher in children exposed to SHS, leading to an increase in OS and to artery dilation [104]. OS may play a major role in the metabolic syndrome, too [105]. It is described that SHS leads to metabolic syndrome in adolescents [106] and children, especially in combination with low intake of vitamin E or omega-3 polyunsaturated fatty acids [107]. Neonates exposed to PTS were shown to have lower levels of adiponectin and higher levels of visfatin, indicating a less beneficial OS profile compared to non-exposed new-borns [108]. In an urban study, adolescents exposed to SHS showed an increase in urinary 15-F2t-isoprostane, a specific product of lipid peroxidation and biomarker for OS level [109].
Kobayashi et al. [110] demonstrated in their study of 2014 that SHS-induced OS reduces the histone deacetylase-2 (HDAC2) function by the activation of phosphoinositide-3-kinase (PI3K), which in turn reduces corticosteroid sensitivity. This corticosteroid insensitiveness make treatment by corticosteroids of severe asthma in children even more difficult [111]. In 2010, Cohen et al. [112] reported that PTS exposure reduces the positive effect of inhaled corticosteroids in asthmatic children. Thus, asthmatic children need higher doses of inhaled corticosteroids for treatment [113].

6. Hormone Status

Several investigations have suggested that PTS exposure leads to hormonal changes in childhood and later life, with increasing risks for adiposity and metabolic and endocrine dysfunction [114]. A higher plasma concentration of the appetite-stimulating hormone ghrelin was shown in 19-year-old young adults exposed to PTS [115]. PTS exposure can influence the foetal endocrine hormone system, like changes in the production of cortisol, oestrogens, androgens, and hormones of the pituitary [116]. PTS and SHS exposure can alter reproductive hormones like luteinizing hormone and inhibin B and, therefore, influence the puberty of girls [117]. Another study showed that daughters exposed to PTS had an earlier age of menarche, but only non-significant lower levels of testosterone and dehydroepiandrosterone-sulphate, and no changes in serum levels of other reproductive hormones [118]. It was found that the positive association between high serum thyrotropin (TSH) levels and body mass index is stronger in SHS-exposed adolescents aged 11–17 than in the unexposed [119]. This boosting effect of SHS exposure was not found in children aged 3–11. Filis et al. [120] reported an association between disturbances of foetal thyroid gland development and maternal smoking and maternal overweight, possibly resulting in influences on endocrine function, foetal metabolism, brain development, cardiac output and adverse post-natal health effects. The hormone leptin will be expressed in adipocytes, regulates food intake and basal metabolism, and correlates with the body mass index with influence on vascular function [121]. In 10-year-old children, SHS exposure was shown to increase leptin plasma concentration [70].

7. Genetic Predisposition

The genetic predisposition may influence the health effects of PTS and SHS exposure in childhood until later life. In the following, different genetic variants of several genes, e.g., glutathione S-transferase genes, interleukin (IL) genes or variants at chromosome 17, are described which may lead to increased adverse health effects (e.g., asthma, wheezing, sudden infant death syndrome, congenital heart defects). Table 2 shows a summary and additional information to the presented genetic predispositions.

7.1. Glutathione S-Transferase (GST) Genes

GSTs, including the isoforms GSTP1, GSTM1, and GSTT1, are multifunctional enzymes for cellular detoxification and are involved in oxidative stress pathways, among others, in the lung [122], and are associated, among others, with asthma and wheezing in children exposed to PTS or SHS [17,18]. In a Swedish prospective birth cohort study (n = 982 wheezers and non-wheezers up to age 4), children with specific variations of three single nucleotide polymorphisms (SNPs) in the GSTP1 gene (Ile105Val, Intron 5, Intron 6) and exposure to SHS had a higher risk of early childhood wheezing, but with no allergic sensitization [123]. In Taiwanese school children (216 wheezers and 185 non-wheezers), homozygosity for GSTP1 Val-105 was significantly associated with both current and ever wheezing, whereas homozygosity for GSTP1 Ile-105 was associated with current wheezing but not with ever wheezing [124]. In a study at 504 children and adolescents with asthma aged between 3 and 21 years, children exposed to SHS and with the GSTP1 polymorphism GG (Val105Val) at nucleotide 1695 or null for the GSTM1 gene were more prone to asthma [125]. A Taiwanese longitudinal birth cohort study (n = 591 children, 138 asthmatics at age 6) found that the GSTM1 null genotype could have a gender-specific effect on the development of asthma. Girls who were not exposed to PTS were more protected against asthma. The GSTM1 null type becomes a risk factor for prenatal exposed boys and girls [126]. Another study in 1124 schoolchildren aged 7–12 years described that SHS-exposed children with the GSTP1 polymorphism AA (Ile105Ile) at nucleotide 1695 showed an increased risk of asthma, too. This effect was boosted by a low intake of vitamin A [127]. At 1132 school children, a common haplotype of GSTP1 (including the polymorphism Ile105Val) was associated with a lower risk of respiratory illness. This effect of protection was lost in those children exposed to PTS or SHS [128].
In contrary to these findings, Turner et al. [129] found no evidence for a significant association between the investigated GST variants GSTP1 Val-105 (n = 3692 children), GSTM1 null and GSTT1 null (n = 2362 children), SHS exposure and asthma attacks in children. The authors concluded that the findings of previous studies, positive associations between GSTT1 null and asthma and GSTM1 null and asthma severity, were false-positive findings.
GST genes may not only be associated with wheezing and asthma. PTS exposure among children with the GSTM1 null genotype was also associated with increased adverse effects on cognition in pre-schoolers [130]. Regarding sudden infant death syndrome (SIDS), a significant association was found between tobacco smoke exposure and GSTM1 null genotype, but no association between GSTT1 null and SIDS [131]. Taiwanese children carrying GSTM1 null and homozygous GSTP1 Ile-105 showed an increasing prevalence of atopic dermatitis when exposed to PTS [132]. Li et al. [133] found an association between congenital heart defects (CHDs) and polymorphisms in GST genes in neonates induced by maternal smoking. Regarding the lung function in adolescents, an atopy cohort study found that exposed children may be more susceptible to lung function impairment in later life if they are homozygous for GSTP1 Ile-105 allele or carriers of the null mutation at GSTM1 or GSTT1 [134].

7.2. Anti-Inflammatory Cytokine Genes

Anti-inflammatory cytokines like tumour necrosis factor-alpha (TNF-α), transforming growth factor-beta (TGF-β) and interleukins (IL) regulate the human immune response by controlling the pro-inflammatory response [30]. In this respect, children with the TNF-308A variant (both homozygous and heterozygous) were more susceptible to respiratory illness by SHS compared to children homozygous for the common TNF-308G allele [135]. SNP variations of TNF (-857C/T, Intron 1, Intron 3) in combination with early maternal smoking interacted with a higher risk of early childhood wheezing but not with allergic sensitization [123].
Regarding the TGF-beta1 gene, it was found that the combination of PTS exposure and the TGF-beta1-509TT genotype of the child increased the risk of childhood asthma [136].
IL-4, IL-5 and IL-13 are associated with asthma, too [31]. It was found in African-American infants exposed to SHS that the CT and TT genotypes for IL-4 C-589T increase the risk of wheezing [137]. In an investigation of the Isle of Wight birth cohort, the common IL 13 haplotype pair CCG/CCG and a single SNP increased the negative effect of maternal smoking during pregnancy on early-onset persistent wheezing and persistent asthma in childhood [138].

7.3. CD14 Gene

CD14 is a receptor for lipopolysaccharide (LPS, endotoxin) on the surface of macrophages, monocytes, and neutrophil blood cells (membrane-anchored = mCD14). CD 14 exists also as a soluble serum protein (sCD14). Both are important molecules regarding endotoxin-dependent signal transduction [139]. It was reported for SHS-exposed children aged four years that an AA genotype in the 3’untranslated region of CD14 was associated with lower IgE levels than in children not exposed. No differences in IgE levels in exposed and non-exposed children were found for the genotype CC/CA [140]. Hussein et al. [141] found that the CD14 genotypes -159TT and -550TT are associated with elevated serum IgE levels in Egyptian children exposed to SHS and may contribute to atopy predisposition.

7.4. Variants at Chromosome 17 Region q21

Bouzigon et al. [142] tested 36 SNPs at chromosome 17q21 for an association with early-onset asthma. They found 11 genetic variants increasing the risk of early-onset asthma, especially at early-life exposure to SHS. These 11 SNPs distributes to four genes: IKZF3 (involved in lymphocyte development), ZPBP2 (zona pellucida-binding protein 2), GSDMB (encodes one gasdermin protein which is involved in skin differentiation and epithelial barrier function) and ORMDL3 (encodes a transmembrane protein). The authors discussed that GSDMB and ORMDL3 in particular may be involved in viral respiratory infections, leading in turn to an increased risk of asthma. The interaction among 17q21variants, SHS exposure of children, and paediatric asthma was confirmed in North Americans of European ancestry, but without the finding of age-of-onset [143]. Both PTS exposure and SHS exposure in early postnatal life and a 17q21 SNP variant increased the asthma-like symptoms in preschoolers [144]. For one SNP on 17q21, an influence on childhood asthma was assumed, but not on asthma risk in adulthood [145]. No association was found for 17q21 variants and late-onset asthma [146].

7.5. ATPase-Related Genes and Bronchial Hyper-Responsiveness

A genetic variant in the DNAH9 gene (chromosome 17p11) in combination with early SHS exposure seems to be linked with bronchial hyper-responsiveness (BHR), an important characteristic for asthma [147,148]. Another two candidate genes, ATP8A1 (chromosome 4) and ABCA1 (chromosome 9), interacting with early-life SHS exposure and BHR, were identified [149].

7.6. Mannose-Binding Lectin-2 (MBL2) Gene

Several genetic polymorphisms and haplotypes in the MBL2 gene increased the lung cancer risk in adults exposed to SHS in their childhood [150]. This is due to the genetic background of MBL2 with consequences in inherent immunity.

7.7. Flavin-Monooxygenase-3 (FMO3) Gene

The common polymorphism G472A of the FMO3 gene may be a risk factor for SIDS in children exposed to PTS [151]. The authors declared their study as the first one demonstrating an interaction between a gene and an environmental factor in SIDS.

7.8. O-Sialoglycoprotein Endopeptidase (OSGEP) Gene

CHDs are among the most prevalent birth defects and are a major cause of death in early childhood [152]. In infants, four SNPs in the OSGEP gene were described as increasing the risk moderately for the occurrence of CHDs in the presence of maternal smoking [153].

7.9. MSX1 Gene

MSX1, a homoeobox gene playing key roles in craniofacial development, is involved in the formation of orofacial clefts (OFC), a common birth defect [154]. The combination of allele 4 homozygosity of the child and PTS exposure increased the risk of OFC [155].

8. Protein Expression in Foetal Liver

In a study on liver samples from aborted foetuses, maternal smoking was gender-specifically associated with changes in the expression pattern of metabolic enzyme transcripts [157]. The authors detected in male foetuses exposed to maternal smoke an expression increase of eight mRNA transcripts (GSTT1, GSTP1, CYP1A1, EPHX1, NQO1, AHR, AS3MT, GLRX2) and a decrease of three transcripts (GGT1, CYP2R1, CAR). In female foetuses, an increase of two transcripts (CYP3A7, EPHX1) was detected. They emphasised the impact on foetal liver activity by environmental toxicants like tobacco smoke. Drake and colleagues [158] showed in foetal livers widespread sex-specific effects by maternal smoking on the expression of key enzymes of the 1-carbon metabolism, level of vitamin B12 and homocysteine in foetal plasma.
Filis et al. [159] described a broad range of dysregulated protein expression in foetal livers induced by maternal smoking in a sex-specific manner, too. Among others, they found expression differences in proteins related to detoxification, homeostasis, protein processing and secretion, necrosis and cancer development, proliferation, apoptosis, and inflammation.

9. Dysregulation of Diverse MicroRNAs

MicroRNAs (miRNAs) are small RNAs with about 22 nucleotides which regulate posttranscriptional gene expression [160]. Placental miRNAs are co-responsible for the development of maternal placenta and foetus. It was ascertained that smoking during pregnancy can down-regulate miRNA-16, miRNA-21, and miRNA-146a in the placenta [161]. On the other side, an up-regulation of miRNA-223 in maternal and cord blood by PTS exposure was noticed to be associated with lower Treg cell numbers in cord blood and succeeding risks for allergies [75]. Lower expression of miR-199a1 in children induced by PTS exposure was reported [162]. This down-regulation leads to lower expression of the receptor-tyrosine-kinase AXL gene and might increase the risks of bronchitic symptoms, especially in combination with higher AXL methylation [162,163]. A recent study showed positive associations between indoor air pollution, e.g., induced by SHS, higher serum levels of miR-155 and asthma in childhood [164].

10. Leukocyte Telomere Length

Telomeres are repetitive DNA sequences. Every cell divide shortens the telomeres. The length of telomeres is associated with the biological age: the shorter the telomere, the higher the biological age [165]. A recent birth cohort study (n = 1396 children aged 5 to 12 years) suggested for the first time that PTS exposure in early life can shorten leukocyte telomeres in children, inducing premature biological ageing, even at an early age [166].

11. DNA Methylation

Tobacco smoke is a potent environmental factor for DNA methylation [167]. PTS exposure can induce epigenetic changes in the foetus with sequelae in later life, among others altering risks for allergic diseases, metabolic diseases, or cardiac disorders [168,169,170]. Many studies focussed on DNA methylations with differences in the global pattern: hyper-methylation in placental tissue and hypo-methylation in cord blood and several types of foetal cell or tissue [171]. Different methylation patterns were found for different placental and foetal genes [74,172,173,174,175,176,177]. Joubert et al. [178] reported in an epigenome-wide analysis of >470,000 individual cytosine-guanine dinucleotide (CpG) sites in cord blood significant methylation changes at 26 CpG sites in 10 genes associated with maternal smoking. That confirmed and extended a study by Markunas and colleagues [179]. The results suggest that changes in DNA methylation triggered by smoking during pregnancy might underlie some consequences of maternal smoking on offspring (e.g., regarding processes at placental and embryonic development).
A recent review article by Zakarya and colleagues presented the epigenetic influences of maternal smoking and the resulting poor effects on the respiratory system of offspring [180]. Den Dekker et al. found 59 differentially methylated regions in the DNA of cord blood altering childhood lung function, some of them additionally associated with childhood asthma or adult COPD [181]. It should also be mentioned that CpG methylation in the gene loci of Forkhead box P3 (FOXP3) and interferon gamma-γ (IFNγ) can be accelerated by SHS exposure [182]. In turn, this can affect the function of T-cells and is associated with asthma. PTS exposure has also been related to lower promoter methylation in the neuropeptide S receptor 1 (NPSR1), significantly associated with asthma [183], and higher AXL gene methylation at birth, increasing bronchitic symptom risk in childhood [162].
In foetal livers, alterations to the methyl donor availability of vitamin B12 were described [158]. This might lead to changes in DNA methylation of the genes IGF2 and NR3C1 (glucocorticoid receptor) induced by maternal smoking with possible consequences on foetal growth and later cardio-metabolic or neuropsychiatric disorders [158].
Finally, it should not go unmentioned that both opioid receptor mu-1 gene (OPRM1) and PTS exposure are factors for the preference for fatty foods [184,185]. It was assumed that PTS exposure is able to epigenetically modify an OPRM1 allele (rs2281617) increasing fat preference [186]. These findings are potential reasons for the described risk of obesity in offspring exposed to PTS [187].

12. Conclusions

Worldwide, many people are still exposed to SHS, especially in public places, at home or in vehicles. Many children will be affected in health by tobacco smoke exposure both before and after birth. The adverse health effects range from prenatal epigenetic changes, triggering of diseases in childhood to health detriment in later life.
This review summarised scientific knowledge of molecular and genetic mechanisms in children induced by PTS and SHS exposure. Tobacco smoke can alter those mechanisms negatively and can cause several diseases. PTS and SHS increase children’s cancer risk, trigger or worsen allergies and asthma in children, and are harmful to their respiratory tract and cardiovascular system. Genetic predisposition may worsen the health effects of exposure. PTS can lead to poor birth outcomes, pregnancy complications, and foetal maldevelopment. It became clear that further research on mechanisms underlying tobacco smoke-induced diseases is necessary, partly because of contradictory study results. Transnational biomonitoring can show the exposition burden by tobacco smoke, especially in children. In this context, the Consortium to Perform Human Biomonitoring on a European Scale (COPHES), funded by the European Union’s Seventh Framework Programme [188,189], and DiMoPEx (Diagnosis, Monitoring and Prevention of Exposure-related Non-Communicable Diseases) by the European Cooperation in Science and Technology [190,191] should be mentioned.
A deeper understanding of health damage by tobacco smoke exposure, which especially affects the child’s developing body, helps the public and policymakers to react adequately with public health interventions, tightening of smoke-free policies, e.g., smoking bans in vehicles with children, health communication campaigns, and behavioural interventions. It is important to emphasise that children in particular should be protected from exposure to tobacco smoke.

Author Contributions

M.B., D.K., G.M.O., D.Q. and D.A.G. contributed substantially to the conception and design of the manuscript. All authors were involved in drafting the manuscript. M.B. wrote the manuscript which was critically reviewed by all authors. D.K. conceptualised the figure. All authors have read and approved the final version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors thank Simona E. Kloft Zitnik for having the idea for such a review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. NIH National Cancer Institute. Secondhand Smoke and Cancer. 2018. Available online: https://www.cancer.gov/about-cancer/causes-prevention/risk/tobacco/second-hand-smoke-fact-sheet (accessed on 16 September 2019).
  2. Protano, C.; Vitali, M. The new danger of thirdhand smoke: Why passive smoking does not stop at secondhand smoke. Environ. Health Perspect. 2011, 119, A422. [Google Scholar] [CrossRef]
  3. ACS American Cancer Society. Health Risks of Secondhand Smoke. 2019. Available online: https://www.cancer.org/cancer/cancer-causes/tobacco-and-cancer/secondhand-smoke.html (accessed on 16 September 2019).
  4. WHO World Health Organization. Global Health Observatory (GHO) Data. Mortality and Burden of Disease from Second-Hand Smoke. 2004. Available online: https://www.who.int/gho/phe/secondhand_smoke/burden/en/ (accessed on 16 September 2019).
  5. Tsai, J.; Homa, D.M.; Gentzke, A.S.; Mahoney, M.; Sharapova, S.R.; Sosnoff, C.S.; Caron, K.T.; Wang, L.; Melstrom, P.C.; Trivers, K.F. Exposure to Secondhand Smoke Among Nonsmokers - United States, 1988–2014. MMWR Morb. Mortal. Wkly. Rep. 2018, 67, 1342–1346. [Google Scholar] [CrossRef] [Green Version]
  6. Besaratinia, A.; Pfeifer, G.P. Second-hand smoke and human lung cancer. Lancet Oncol. 2008, 9, 657–666. [Google Scholar] [CrossRef] [Green Version]
  7. Lee, P.N.; Hamling, J.S. Environmental tobacco smoke exposure and risk of breast cancer in nonsmoking women. An updated review and meta-analysis. Inhal. Toxicol. 2016, 28, 431–454. [Google Scholar] [CrossRef] [Green Version]
  8. Farber, H.J.; Groner, J.; Walley, S.; Nelson, K. Protecting Children From Tobacco, Nicotine, and Tobacco Smoke. Pediatrics 2015, 136, e1439–e1467. [Google Scholar] [CrossRef] [Green Version]
  9. DiGiacomo, S.I.; Jazayeri, M.A.; Barua, R.S.; Ambrose, J.A. Environmental Tobacco Smoke and Cardiovascular Disease. Int. J. Environ. Res. Public Health 2018, 16, 96. [Google Scholar] [CrossRef] [Green Version]
  10. Strzelak, A.; Ratajczak, A.; Adamiec, A.; Feleszko, W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. Int. J. Environ. Res. Public Health 2018, 15, 1033. [Google Scholar] [CrossRef] [Green Version]
  11. Rahman, I.; MacNee, W. Lung glutathione and oxidative stress: Implications in cigarette smoke-induced airway disease. Am. J. Physiol. 1999, 277, L1067–L1088. [Google Scholar] [CrossRef]
  12. Han, C.; Liu, Y.; Gong, X.; Ye, X.; Zhou, J. Relationship Between Secondhand Smoke Exposure and Depressive Symptoms: A Systematic Review and Dose(-)Response Meta-Analysis. Int. J. Environ. Res. Public Health 2019, 16, 1356. [Google Scholar] [CrossRef] [Green Version]
  13. Mund, M.; Louwen, F.; Klingelhoefer, D.; Gerber, A. Smoking and pregnancy—A review on the first major environmental risk factor of the unborn. Int. J. Environ. Res. Public Health 2013, 10, 6485–6499. [Google Scholar] [CrossRef] [Green Version]
  14. Waterland, R.A.; Michels, K.B. Epigenetic epidemiology of the developmental origins hypothesis. Annu. Rev. Nutr. 2007, 27, 363–388. [Google Scholar] [CrossRef]
  15. U.S. Department of Health and Human Services. The Health Consequences of Smoking—50 Years of Progress; A Report of the Surgeon General; U.S. Department of Health and Human Services, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health: Atlanta, GA, USA, 2014. Available online: https://www.ncbi.nlm.nih.gov/books/NBK179276/pdf/Bookshelf_NBK179276.pdf (accessed on 17 September 2019).
  16. von Ehrenstein, O.S.; von Mutius, E.; Maier, E.; Hirsch, T.; Carr, D.; Schaal, W.; Roscher, A.A.; Olgemoller, B.; Nicolai, T.; Weiland, S.K. Lung function of school children with low levels of alpha1-antitrypsin and tobacco smoke exposure. Eur. Respir. J. 2002, 19, 1099–1106. [Google Scholar] [CrossRef]
  17. Gilliland, F.D.; Li, Y.F.; Dubeau, L.; Berhane, K.; Avol, E.; McConnell, R.; Gauderman, W.J.; Peters, J.M. Effects of glutathione S-transferase M1, maternal smoking during pregnancy, and environmental tobacco smoke on asthma and wheezing in children. Am. J. Respir. Crit. Care Med. 2002, 166, 457–463. [Google Scholar] [CrossRef] [Green Version]
  18. Kabesch, M.; Hoefler, C.; Carr, D.; Leupold, W.; Weiland, S.K.; von Mutius, E. Glutathione S transferase deficiency and passive smoking increase childhood asthma. Thorax 2004, 59, 569–573. [Google Scholar] [CrossRef] [Green Version]
  19. Chen, Y.; Wong, G.W.; Li, J. Environmental Exposure and Genetic Predisposition as Risk Factors for Asthma in China. Allergy Asthma Immunol. Res. 2016, 8, 92–100. [Google Scholar] [CrossRef] [Green Version]
  20. Oberg, M.; Jaakkola, M.S.; Woodward, A.; Peruga, A.; Pruss-Ustun, A. Worldwide burden of disease from exposure to second-hand smoke: A retrospective analysis of data from 192 countries. Lancet 2011, 377, 139–146. [Google Scholar] [CrossRef]
  21. Homa, D.M.; Neff, L.J.; King, B.A.; Caraballo, R.S.; Bunnell, R.E.; Babb, S.D.; Garrett, B.E.; Sosnoff, C.S.; Wang, L.; Centers for Disease, C.; et al. Vital signs: Disparities in nonsmokers’ exposure to secondhand smoke--United States, 1999–2012. MMWR Morb. Mortal. Wkly. Rep. 2015, 64, 103–108. [Google Scholar]
  22. Lupsa, I.R.; Nunes, B.; Ligocka, D.; Gurzau, A.E.; Jakubowski, M.; Casteleyn, L.; Aerts, D.; Biot, P.; Den Hond, E.; Castano, A.; et al. Urinary cotinine levels and environmental tobacco smoke in mothers and children of Romania, Portugal and Poland within the European human biomonitoring pilot study. Environ. Res. 2015, 141, 106–117. [Google Scholar] [CrossRef]
  23. WHO World Health Organization. Tobacco. Key Facts. 2019. Available online: https://www.who.int/news-room/fact-sheets/detail/tobacco (accessed on 17 September 2019).
  24. Lau, E.M.; Celermajer, D.S. Protecting our children from environmental tobacco smoke: One of our great healthcare challenges. Eur. Heart J. 2014, 35, 2452–2453. [Google Scholar] [CrossRef] [Green Version]
  25. Van den Steen, P.E.; Dubois, B.; Nelissen, I.; Rudd, P.M.; Dwek, R.A.; Opdenakker, G. Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9). Crit. Rev. Biochem. Mol. 2002, 37, 375–536. [Google Scholar] [CrossRef]
  26. De, S.; Leong, S.C.; Fenton, J.E.; Carter, S.D.; Clarke, R.W.; Jones, A.S. The effect of passive smoking on the levels of matrix metalloproteinase 9 in nasal secretions of children. Am. J. Rhinol. Allergy 2011, 25, 226–230. [Google Scholar] [CrossRef] [PubMed]
  27. Vignola, A.M.; Riccobono, L.; Mirabella, A.; Profita, M.; Chanez, P.; Bellia, V.; Mautino, G.; D’Accardi, P.; Bousquet, J.; Bonsignore, G. Sputum metalloproteinase-9/tissue inhibitor of metalloproteinase-1 ratio correlates with airflow obstruction in asthma and chronic bronchitis. Am. J. Respir. Crit. Care Med. 1998, 158, 1945–1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Kopp, B.T.; Thompson, R.; Kim, J.; Konstan, R.; Diaz, A.; Smith, B.; Shrestha, C.; Rogers, L.K.; Hayes, D., Jr.; Tumin, D.; et al. Secondhand smoke alters arachidonic acid metabolism and inflammation in infants and children with cystic fibrosis. Thorax 2019, 74, 237–246. [Google Scholar] [CrossRef]
  29. Yilmaz, O.; Turkeli, A.; Onur, E.; Bilge, S.; Yuksel, H. Secondhand tobacco smoke and severity in wheezing children: Nasal oxidant stress and inflammation. J. Asthma 2018, 55, 477–482. [Google Scholar] [CrossRef]
  30. Opal, S.M.; DePalo, V.A. Anti-inflammatory cytokines. Chest 2000, 117, 1162–1172. [Google Scholar] [CrossRef] [Green Version]
  31. Lambrecht, B.N.; Hammad, H.; Fahy, J.V. The Cytokines of Asthma. Immunity 2019, 50, 975–991. [Google Scholar] [CrossRef]
  32. Berry, M.; Brightling, C.; Pavord, I.; Wardlaw, A. TNF-alpha in asthma. Curr. Opin. Pharmacol. 2007, 7, 279–282. [Google Scholar] [CrossRef] [Green Version]
  33. Chahal, N.; McLain, A.C.; Ghassabian, A.; Michels, K.A.; Bell, E.M.; Lawrence, D.A.; Yeung, E.H. Maternal Smoking and Newborn Cytokine and Immunoglobulin Levels. Nicotine Tob. Res. 2017, 19, 789–796. [Google Scholar] [CrossRef] [Green Version]
  34. Wilson, K.M.; Wesgate, S.C.; Pier, J.; Weis, E.; Love, T.; Evans, K.; Chhibber, A. Secondhand smoke exposure and serum cytokine levels in healthy children. Cytokine 2012, 60, 34–37. [Google Scholar] [CrossRef] [Green Version]
  35. Mahabee-Gittens, E.M.; Merianos, A.L.; Fulkerson, P.C.; Stone, L.; Matt, G.E. The Association of Environmental Tobacco Smoke Exposure and Inflammatory Markers in Hospitalized Children. Int. J. Environ. Res. Public Health 2019, 16, 4625. [Google Scholar] [CrossRef] [Green Version]
  36. Riis, J.L.; Granger, D.A.; DiPietro, J.A.; Bandeen-Roche, K.; Johnson, S.B. Salivary cytokines as a minimally-invasive measure of immune functioning in young children: Correlates of individual differences and sensitivity to laboratory stress. Dev. Psychobiol. 2015, 57, 153–167. [Google Scholar] [CrossRef] [PubMed]
  37. Feleszko, W.; Zawadzka-Krajewska, A.; Matysiak, K.; Lewandowska, D.; Peradzynska, J.; Dinh, Q.T.; Hamelmann, E.; Groneberg, D.A.; Kulus, M. Parental tobacco smoking is associated with augmented IL-13 secretion in children with allergic asthma. J. Allergy Clin. Immunol. 2006, 117, 97–102. [Google Scholar] [CrossRef] [PubMed]
  38. Laitinen, L.A.; Laitinen, A.; Haahtela, T.; Vilkka, V.; Spur, B.W.; Lee, T.H. Leukotriene E4 and granulocytic infiltration into asthmatic airways. Lancet 1993, 341, 989–990. [Google Scholar] [CrossRef]
  39. Kumlin, M. Measurement of leukotrienes in humans. Am. J. Respir. Crit. Care Med. 2000, 161, S102–S106. [Google Scholar] [CrossRef]
  40. Lazarus, S.C.; Chinchilli, V.M.; Rollings, N.J.; Boushey, H.A.; Cherniack, R.; Craig, T.J.; Deykin, A.; DiMango, E.; Fish, J.E.; Ford, J.G.; et al. Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma. Am. J. Respir. Crit. Care Med. 2007, 175, 783–790. [Google Scholar] [CrossRef]
  41. Rabinovitch, N.; Strand, M.; Stuhlman, K.; Gelfand, E.W. Exposure to tobacco smoke increases leukotriene E4-related albuterol usage and response to montelukast. J. Allergy Clin. Immunol. 2008, 121, 1365–1371. [Google Scholar] [CrossRef]
  42. Rabinovitch, N.; Reisdorph, N.; Silveira, L.; Gelfand, E.W. Urinary leukotriene E(4) levels identify children with tobacco smoke exposure at risk for asthma exacerbation. J. Allergy Clin. Immunol. 2011, 128, 323–327. [Google Scholar] [CrossRef] [Green Version]
  43. Kott, K.S.; Salt, B.H.; McDonald, R.J.; Jhawar, S.; Bric, J.M.; Joad, J.P. Effect of secondhand cigarette smoke, RSV bronchiolitis and parental asthma on urinary cysteinyl LTE4. Pediatr. Pulmonol. 2008, 43, 760–766. [Google Scholar] [CrossRef]
  44. Chilmonczyk, B.A.; Salmun, L.M.; Megathlin, K.N.; Neveux, L.M.; Palomaki, G.E.; Knight, G.J.; Pulkkinen, A.J.; Haddow, J.E. Association between exposure to environmental tobacco smoke and exacerbations of asthma in children. N. Engl. J. Med. 1993, 328, 1665–1669. [Google Scholar] [CrossRef]
  45. Lang, J.E.; Dozor, A.J.; Holbrook, J.T.; Mougey, E.; Krishnan, S.; Sweeten, S.; Wise, R.A.; Teague, W.G.; Wei, C.Y.; Shade, D.; et al. Biologic mechanisms of environmental tobacco smoke in children with poorly controlled asthma: Results from a multicenter clinical trial. J. Allergy Clin. Immunol. Pract. 2013, 1, 172–180. [Google Scholar] [CrossRef] [Green Version]
  46. Hernandez-Alvidrez, E.; Alba-Reyes, G.; Munoz-Cedillo, B.C.; Arreola-Ramirez, J.L.; Furuya, M.E.; Becerril-Angeles, M.; Vargas, M.H. Passive smoking induces leukotriene production in children: Influence of asthma. J. Asthma 2013, 50, 347–353. [Google Scholar] [CrossRef]
  47. Gill, R.; Krishnan, S.; Dozor, A.J. Low-level environmental tobacco smoke exposure and inflammatory biomarkers in children with asthma. J. Asthma 2014, 51, 355–359. [Google Scholar] [CrossRef]
  48. Marmarinos, A.; Saxoni-Papageorgiou, P.; Cassimos, D.; Manoussakis, E.; Tsentidis, C.; Doxara, A.; Paraskakis, I.; Gourgiotis, D. Urinary leukotriene E4 levels in atopic and non-atopic preschool children with recurrent episodic (viral) wheezing: A potential marker? J. Asthma 2015, 52, 554–559. [Google Scholar] [CrossRef]
  49. Chiu, C.Y.; Tsai, M.H.; Yao, T.C.; Tu, Y.L.; Hua, M.C.; Yeh, K.W.; Huang, J.L. Urinary LTE4 levels as a diagnostic marker for IgE-mediated asthma in preschool children: A birth cohort study. PLoS ONE 2014, 9, e115216. [Google Scholar] [CrossRef] [Green Version]
  50. Hagan, J.B.; Laidlaw, T.M.; Divekar, R.; O’Brien, E.K.; Kita, H.; Volcheck, G.W.; Hagan, C.R.; Lal, D.; Teaford, H.G., 3rd; Erwin, P.J.; et al. Urinary Leukotriene E4 to Determine Aspirin Intolerance in Asthma: A Systematic Review and Meta-Analysis. J. Allergy Clin. Immunol. Pract. 2017, 5, 990–997. [Google Scholar] [CrossRef]
  51. Hoffman, B.C.; Rabinovitch, N. Urinary Leukotriene E4 as a Biomarker of Exposure, Susceptibility, and Risk in Asthma: An Update. Immunol. Allergy Clin. 2018, 38, 599–610. [Google Scholar] [CrossRef]
  52. Taal, H.R.; Geelhoed, J.J.; Steegers, E.A.; Hofman, A.; Moll, H.A.; Lequin, M.; van der Heijden, A.J.; Jaddoe, V.W. Maternal smoking during pregnancy and kidney volume in the offspring: The Generation R Study. Pediatr. Nephrol. 2011, 26, 1275–1283. [Google Scholar] [CrossRef] [Green Version]
  53. Hogan, S.L.; Vupputuri, S.; Guo, X.; Cai, J.; Colindres, R.E.; Heiss, G.; Coresh, J. Association of cigarette smoking with albuminuria in the United States: The third National Health and Nutrition Examination Survey. Ren. Fail. 2007, 29, 133–142. [Google Scholar] [CrossRef]
  54. Omoloja, A.; Chand, D.; Greenbaum, L.; Wilson, A.; Bastian, V.; Ferris, M.; Bernert, J.; Stolfi, A.; Patel, H. Cigarette smoking and second-hand smoking exposure in adolescents with chronic kidney disease: A study from the Midwest Pediatric Nephrology Consortium. Nephrol. Dial. Transplant. 2011, 26, 908–913. [Google Scholar] [CrossRef] [Green Version]
  55. Garcia-Esquinas, E.; Loeffler, L.F.; Weaver, V.M.; Fadrowski, J.J.; Navas-Acien, A. Kidney function and tobacco smoke exposure in US adolescents. Pediatrics 2013, 131, e1415–e1423. [Google Scholar] [CrossRef] [Green Version]
  56. Omoloja, A.; Jerry-Fluker, J.; Ng, D.K.; Abraham, A.G.; Furth, S.; Warady, B.A.; Mitsnefes, M. Secondhand smoke exposure is associated with proteinuria in children with chronic kidney disease. Pediatr. Nephrol. 2013, 28, 1243–1251. [Google Scholar] [CrossRef] [Green Version]
  57. Kooijman, M.N.; Bakker, H.; Franco, O.H.; Hofman, A.; Taal, H.R.; Jaddoe, V.W. Fetal Smoke Exposure and Kidney Outcomes in School-Aged Children. Am. J. Kidney Dis. 2015, 66, 412–420. [Google Scholar] [CrossRef]
  58. He, J.; Vupputuri, S.; Allen, K.; Prerost, M.R.; Hughes, J.; Whelton, P.K. Passive smoking and the risk of coronary heart disease—A meta-analysis of epidemiologic studies. N. Engl. J. Med. 1999, 340, 920–926. [Google Scholar] [CrossRef]
  59. Torok, J.; Gvozdjakova, A.; Kucharska, J.; Balazovjech, I.; Kysela, S.; Simko, F.; Gvozdjak, J. Passive smoking impairs endothelium-dependent relaxation of isolated rabbit arteries. Physiol. Res. 2000, 49, 135–141. [Google Scholar]
  60. Hackshaw, A.; Rodeck, C.; Boniface, S. Maternal smoking in pregnancy and birth defects: A systematic review based on 173 687 malformed cases and 11.7 million controls. Hum. Reprod. Update 2011, 17, 589–604. [Google Scholar] [CrossRef] [Green Version]
  61. Groner, J.A.; Huang, H.; Nagaraja, H.; Kuck, J.; Bauer, J.A. Secondhand smoke exposure and endothelial stress in children and adolescents. Acad. Pediatr. 2015, 15, 54–60. [Google Scholar] [CrossRef] [Green Version]
  62. Raitakari, O.T.; Juonala, M.; Kahonen, M.; Taittonen, L.; Laitinen, T.; Maki-Torkko, N.; Jarvisalo, M.J.; Uhari, M.; Jokinen, E.; Ronnemaa, T.; et al. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: The Cardiovascular Risk in Young Finns Study. JAMA 2003, 290, 2277–2283. [Google Scholar] [CrossRef]
  63. Gunes, T.; Koklu, E.; Yikilmaz, A.; Ozturk, M.A.; Akcakus, M.; Kurtoglu, S.; Coskun, A.; Koklu, S. Influence of maternal smoking on neonatal aortic intima-media thickness, serum IGF-I and IGFBP-3 levels. Eur. J. Pediatr. 2007, 166, 1039–1044. [Google Scholar] [CrossRef]
  64. Geerts, C.C.; Bots, M.L.; van der Ent, C.K.; Grobbee, D.E.; Uiterwaal, C.S. Parental smoking and vascular damage in their 5-year-old children. Pediatrics 2012, 129, 45–54. [Google Scholar] [CrossRef] [Green Version]
  65. Gall, S.; Huynh, Q.L.; Magnussen, C.G.; Juonala, M.; Viikari, J.S.; Kahonen, M.; Dwyer, T.; Raitakari, O.T.; Venn, A. Exposure to parental smoking in childhood or adolescence is associated with increased carotid intima-media thickness in young adults: Evidence from the Cardiovascular Risk in Young Finns study and the Childhood Determinants of Adult Health Study. Eur. Heart J. 2014, 35, 2484–2491. [Google Scholar] [CrossRef]
  66. Groh, C.A.; Vittinghoff, E.; Benjamin, E.J.; Dupuis, J.; Marcus, G.M. Childhood Tobacco Smoke Exposure and Risk of Atrial Fibrillation in Adulthood. J. Am. Coll. Cardiol. 2019, 74, 1658–1664. [Google Scholar] [CrossRef] [PubMed]
  67. Black, S.; Kushner, I.; Samols, D. C-reactive Protein. J. Biol. Chem. 2004, 279, 48487–48490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Cook, D.G.; Mendall, M.A.; Whincup, P.H.; Carey, I.M.; Ballam, L.; Morris, J.E.; Miller, G.J.; Strachan, D.P. C-reactive protein concentration in children: Relationship to adiposity and other cardiovascular risk factors. Atherosclerosis 2000, 149, 139–150. [Google Scholar] [CrossRef]
  69. Wilkinson, J.D.; Lee, D.J.; Arheart, K.L. Secondhand smoke exposure and C-reactive protein levels in youth. Nicotine Tob. Res. 2007, 9, 305–307. [Google Scholar] [CrossRef]
  70. Nagel, G.; Arnold, F.J.; Wilhelm, M.; Link, B.; Zoellner, I.; Koenig, W. Environmental tobacco smoke and cardiometabolic risk in young children: Results from a survey in south-west Germany. Eur. Heart J. 2009, 30, 1885–1893. [Google Scholar] [CrossRef] [Green Version]
  71. Kang, E.; Kim, S.Y.; Chang, S.S.; Lim, S.; Kim, H.C.; Lee, C.G.; Kim, Y.M.; Kim, S.Y.; Lee, K.J.; Kim, S.; et al. Environmental Tobacco Smoke Exposure at Home and High-Sensitivity C-Reactive Protein Levels in Three-to-Five-Year-Old Children. Int. J. Environ. Res. Public Health 2017, 14, 1105. [Google Scholar] [CrossRef] [Green Version]
  72. Wang, D.; Juonala, M.; Viikari, J.S.A.; Wu, F.; Hutri-Kahonen, N.; Raitakari, O.T.; Magnussen, C.G. Exposure to Parental Smoking in Childhood is Associated with High C-Reactive Protein in Adulthood: The Cardiovascular Risk in Young Finns Study. J. Atheroscler. Thromb. 2017, 24, 1231–1241. [Google Scholar] [CrossRef] [Green Version]
  73. Palomares, O.; Yaman, G.; Azkur, A.K.; Akkoc, T.; Akdis, M.; Akdis, C.A. Role of Treg in immune regulation of allergic diseases. Eur. J. Immunol. 2010, 40, 1232–1240. [Google Scholar] [CrossRef]
  74. Hinz, D.; Bauer, M.; Roder, S.; Olek, S.; Huehn, J.; Sack, U.; Borte, M.; Simon, J.C.; Lehmann, I.; Herberth, G.; et al. Cord blood Tregs with stable FOXP3 expression are influenced by prenatal environment and associated with atopic dermatitis at the age of one year. Allergy 2012, 67, 380–389. [Google Scholar] [CrossRef]
  75. Herberth, G.; Bauer, M.; Gasch, M.; Hinz, D.; Roder, S.; Olek, S.; Kohajda, T.; Rolle-Kampczyk, U.; von Bergen, M.; Sack, U.; et al. Maternal and cord blood miR-223 expression associates with prenatal tobacco smoke exposure and low regulatory T-cell numbers. J. Allergy Clin. Immunol. 2014, 133, 543–550. [Google Scholar] [CrossRef]
  76. Hedrick, S.M.; Hess Michelini, R.; Doedens, A.L.; Goldrath, A.W.; Stone, E.L. FOXO transcription factors throughout T cell biology. Nat. Rev. Immunol. 2012, 12, 649–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lee, J.; Kim, C.J.; Kim, J.S.; Lee, D.C.; Ahn, S.; Yoon, B.H. Increased miR-223 expression in foetal organs is a signature of acute chorioamnionitis with systemic consequences. J. Cell. Mol. Med. 2018, 22, 1179–1189. [Google Scholar] [CrossRef] [PubMed]
  78. Jing, W.; Wang, W.; Liu, Q. Passive smoking induces pediatric asthma by affecting the balance of Treg/Th17 cells. Pediatr. Res. 2019, 85, 469–476. [Google Scholar] [CrossRef] [PubMed]
  79. Shi, Y.H.; Shi, G.C.; Wan, H.Y.; Jiang, L.H.; Ai, X.Y.; Zhu, H.X.; Tang, W.; Ma, J.Y.; Jin, X.Y.; Zhang, B.Y. Coexistence of Th1/Th2 and Th17/Treg imbalances in patients with allergic asthma. Chin. Med. J. 2011, 124, 1951–1956. [Google Scholar] [PubMed]
  80. Vardavas, C.I.; Plada, M.; Tzatzarakis, M.; Marcos, A.; Warnberg, J.; Gomez-Martinez, S.; Breidenassel, C.; Gonzalez-Gross, M.; Tsatsakis, A.M.; Saris, W.H.; et al. Passive smoking alters circulating naive/memory lymphocyte T-cell subpopulations in children. Pediatr. Allergy Immunol. 2010, 21, 1171–1178. [Google Scholar] [CrossRef] [Green Version]
  81. Marseglia, G.L.; Avanzini, M.A.; Caimmi, S.; Caimmi, D.; Marseglia, A.; Valsecchi, C.; Poddighe, D.; Ciprandi, G.; Pagella, F.; Klersy, C.; et al. Passive exposure to smoke results in defective interferon-gamma production by adenoids in children with recurrent respiratory infections. J. Interferon Cytokine Res. 2009, 29, 427–432. [Google Scholar] [CrossRef]
  82. Tagliacarne, S.C.; Valsecchi, C.; Castellazzi, A.M.; Licari, A.; Klersy, C.; Montagna, L.; Castagnoli, R.; Benazzo, M.; Ciprandi, G.; Marseglia, G.L. Impact of passive smoke and/or atopy on adenoid immunoglobulin production in children. Immunol. Lett. 2015, 165, 70–77. [Google Scholar] [CrossRef]
  83. Yao, T.C.; Chang, S.W.; Hua, M.C.; Liao, S.L.; Tsai, M.H.; Lai, S.H.; Tseng, Y.L.; Yeh, K.W.; Tsai, H.J.; Huang, J.L.; et al. Tobacco smoke exposure and multiplexed immunoglobulin E sensitization in children: A population-based study. Allergy 2016, 71, 90–98. [Google Scholar] [CrossRef] [Green Version]
  84. Thacher, J.D.; Gruzieva, O.; Pershagen, G.; Neuman, A.; van Hage, M.; Wickman, M.; Kull, I.; Melen, E.; Bergstrom, A. Parental smoking and development of allergic sensitization from birth to adolescence. Allergy 2016, 71, 239–248. [Google Scholar] [CrossRef] [Green Version]
  85. Feleszko, W.; Ruszczynski, M.; Jaworska, J.; Strzelak, A.; Zalewski, B.M.; Kulus, M. Environmental tobacco smoke exposure and risk of allergic sensitisation in children: A systematic review and meta-analysis. Arch. Dis. Child. 2014, 99, 985–992. [Google Scholar] [CrossRef]
  86. Hirata, K.; Yamano, Y.; Suzuki, H.; Miyagawa, S.; Nakadate, T. Passive smoking is associated with lower serum HDL-C levels in school children. Pediatr. Int. 2010, 52, 252–256. [Google Scholar] [CrossRef] [PubMed]
  87. El-Hodhod, M.A.; Hamdy, A.M.; Ahmed, M.B.; Youssef, S.R.; Aly, S.M. Effect of passive smoking on blood lymphocyte apoptosis in children. Eur. J. Clin. Investig. 2011, 41, 387–392. [Google Scholar] [CrossRef] [PubMed]
  88. Xie, B.; Palmer, P.H.; Pang, Z.; Sun, P.; Duan, H.; Johnson, C.A. Environmental tobacco use and indicators of metabolic syndrome in Chinese adults. Nicotine Tob. Res. 2010, 12, 198–206. [Google Scholar] [CrossRef] [PubMed]
  89. Groner, J.A.; Huang, H.; Joshi, M.S.; Eastman, N.; Nicholson, L.; Bauer, J.A. Secondhand Smoke Exposure and Preclinical Markers of Cardiovascular Risk in Toddlers. J. Pediatr. 2017, 189, 155–161. [Google Scholar] [CrossRef] [Green Version]
  90. Ayer, J.G.; Belousova, E.; Harmer, J.A.; David, C.; Marks, G.B.; Celermajer, D.S. Maternal cigarette smoking is associated with reduced high-density lipoprotein cholesterol in healthy 8-year-old children. Eur. Heart J. 2011, 32, 2446–2453. [Google Scholar] [CrossRef]
  91. Le-Ha, C.; Beilin, L.J.; Burrows, S.; Huang, R.C.; Oddy, W.H.; Hands, B.; Mori, T.A. Gender difference in the relationship between passive smoking exposure and HDL-cholesterol levels in late adolescence. J. Clin. Endocrinol. Metab. 2013, 98, 2126–2135. [Google Scholar] [CrossRef] [Green Version]
  92. Kallio, K.; Jokinen, E.; Saarinen, M.; Hamalainen, M.; Volanen, I.; Kaitosaari, T.; Ronnemaa, T.; Viikari, J.; Raitakari, O.T.; Simell, O. Arterial intima-media thickness, endothelial function, and apolipoproteins in adolescents frequently exposed to tobacco smoke. Circ. Cardiovasc. Qual. Outcomes 2010, 3, 196–203. [Google Scholar] [CrossRef] [Green Version]
  93. Cupul-Uicab, L.A.; Skjaerven, R.; Haug, K.; Travlos, G.S.; Wilson, R.E.; Eggesbo, M.; Hoppin, J.A.; Whitworth, K.W.; Longnecker, M.P. Exposure to tobacco smoke in utero and subsequent plasma lipids, ApoB, and CRP among adult women in the MoBa cohort. Environ. Health Perspect. 2012, 120, 1532–1537. [Google Scholar] [CrossRef] [Green Version]
  94. Rolle-Kampczyk, U.E.; Krumsiek, J.; Otto, W.; Roder, S.W.; Kohajda, T.; Borte, M.; Theis, F.; Lehmann, I.; von Bergen, M. Metabolomics reveals effects of maternal smoking on endogenous metabolites from lipid metabolism in cord blood of newborns. Metabolomics 2016, 12, 76. [Google Scholar] [CrossRef] [Green Version]
  95. Zakhar, J.; Amrock, S.M.; Weitzman, M. Passive and Active Tobacco Exposure and Children’s Lipid Profiles. Nicotine Tob. Res. 2016, 18, 982–987. [Google Scholar] [CrossRef] [Green Version]
  96. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Halliwell, B. Free radicals, antioxidants, and human disease: Curiosity, cause, or consequence? Lancet 1994, 344, 721–724. [Google Scholar] [CrossRef]
  98. Sahiner, U.M.; Birben, E.; Erzurum, S.; Sackesen, C.; Kalayci, O. Oxidative stress in asthma: Part of the puzzle. Pediatr. Allergy Immunol. 2018, 29, 789–800. [Google Scholar] [CrossRef] [PubMed]
  99. Raghuveer, G.; White, D.A.; Hayman, L.L.; Woo, J.G.; Villafane, J.; Celermajer, D.; Ward, K.D.; de Ferranti, S.D.; Zachariah, J.; American Heart Association Committee on Atherosclerosis, Hypertension, and Obesity in the Young of the Council on Cardiovascular Disease in the Young; et al. Cardiovascular Consequences of Childhood Secondhand Tobacco Smoke Exposure: Prevailing Evidence, Burden, and Racial and Socioeconomic Disparities: A Scientific Statement From the American Heart Association. Circulation 2016, 134, e336–e359. [Google Scholar] [CrossRef] [Green Version]
  100. Perrone, S.; Tataranno, M.L.; Stazzoni, G.; Buonocore, G. Biomarkers of oxidative stress in fetal and neonatal diseases. J. Matern. Fetal Neonatal Med. 2012, 25, 2575–2578. [Google Scholar] [CrossRef]
  101. Bono, R.; Bellisario, V.; Romanazzi, V.; Pirro, V.; Piccioni, P.; Pazzi, M.; Bugiani, M.; Vincenti, M. Oxidative stress in adolescent passive smokers living in urban and rural environments. Int. J. Hyg. Environ. Health 2014, 217, 287–293. [Google Scholar] [CrossRef]
  102. Squillacioti, G.; Bellisario, V.; Grignani, E.; Mengozzi, G.; Bardaglio, G.; Dalmasso, P.; Bono, R. The Asti Study: The Induction of Oxidative Stress in A Population of Children According to Their Body Composition and Passive Tobacco Smoking Exposure. Int. J. Environ. Res. Public Health 2019, 16, 490. [Google Scholar] [CrossRef] [Green Version]
  103. Cahill-Smith, S.; Li, J.M. Oxidative stress, redox signalling and endothelial dysfunction in ageing-related neurodegenerative diseases: A role of NADPH oxidase 2. Br. J. Clin. Pharmacol. 2014, 78, 441–453. [Google Scholar] [CrossRef] [Green Version]
  104. Loffredo, L.; Zicari, A.M.; Occasi, F.; Perri, L.; Carnevale, R.; Angelico, F.; Del Ben, M.; Martino, F.; Nocella, C.; De Castro, G.; et al. Role of NADPH oxidase-2 and oxidative stress in children exposed to passive smoking. Thorax 2018, 73, 986–988. [Google Scholar] [CrossRef]
  105. Roberts, C.K.; Sindhu, K.K. Oxidative stress and metabolic syndrome. Life Sci. 2009, 84, 705–712. [Google Scholar] [CrossRef]
  106. Weitzman, M.; Cook, S.; Auinger, P.; Florin, T.A.; Daniels, S.; Nguyen, M.; Winickoff, J.P. Tobacco smoke exposure is associated with the metabolic syndrome in adolescents. Circulation 2005, 112, 862–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Moore, B.F.; Clark, M.L.; Bachand, A.; Reynolds, S.J.; Nelson, T.L.; Peel, J.L. Interactions Between Diet and Exposure to Secondhand Smoke on Metabolic Syndrome Among Children: NHANES 2007–2010. J. Clin. Endocrinol. Metab. 2016, 101, 52–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Chelchowska, M.; Ambroszkiewicz, J.; Gajewska, J.; Rowicka, G.; Maciejewski, T.M.; Mazur, J. Cord Blood Adiponectin and Visfatin Concentrations in relation to Oxidative Stress Markers in Neonates Exposed and Nonexposed In Utero to Tobacco Smoke. Oxid. Med. Cell. Longev. 2016, 2016, 4569108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Bono, R.; Tassinari, R.; Bellisario, V.; Gilli, G.; Pazzi, M.; Pirro, V.; Mengozzi, G.; Bugiani, M.; Piccioni, P. Urban air and tobacco smoke as conditions that increase the risk of oxidative stress and respiratory response in youth. Environ. Res. 2015, 137, 141–146. [Google Scholar] [CrossRef]
  110. Kobayashi, Y.; Bossley, C.; Gupta, A.; Akashi, K.; Tsartsali, L.; Mercado, N.; Barnes, P.J.; Bush, A.; Ito, K. Passive smoking impairs histone deacetylase-2 in children with severe asthma. Chest 2014, 145, 305–312. [Google Scholar] [CrossRef] [Green Version]
  111. Rathkopf, M.M. Passive smoking impairs histone deacetylase-2 in children with severe asthma. Pediatrics 2014, 134 (Suppl. 3), S147–S148. [Google Scholar] [CrossRef] [Green Version]
  112. Cohen, R.T.; Raby, B.A.; Van Steen, K.; Fuhlbrigge, A.L.; Celedon, J.C.; Rosner, B.A.; Strunk, R.C.; Zeiger, R.S.; Weiss, S.T.; Childhood Asthma Management Program Research Group. In utero smoke exposure and impaired response to inhaled corticosteroids in children with asthma. J. Allergy Clin. Immunol. 2010, 126, 491–497. [Google Scholar] [CrossRef] [Green Version]
  113. Podlecka, D.; Malewska-Kaczmarek, K.; Jerzynska, J.; Stelmach, W.; Stelmach, I. Secondhand smoke exposure increased the need for inhaled corticosteroids in children with asthma. Ann. Allergy Asthma Immunol. 2018, 121, 119–121. [Google Scholar] [CrossRef]
  114. Lisboa, P.C.; de Oliveira, E.; de Moura, E.G. Obesity and endocrine dysfunction programmed by maternal smoking in pregnancy and lactation. Front. Physiol. 2012, 3, 437. [Google Scholar] [CrossRef] [Green Version]
  115. Paslakis, G.; Buchmann, A.F.; Westphal, S.; Banaschewski, T.; Hohm, E.; Zimmermann, U.S.; Laucht, M.; Deuschle, M. Intrauterine exposure to cigarette smoke is associated with increased ghrelin concentrations in adulthood. Neuroendocrinology 2014, 99, 123–129. [Google Scholar] [CrossRef]
  116. Duskova, M.; Hruskovicova, H.; Simunkova, K.; Starka, L.; Parizek, A. The effects of smoking on steroid metabolism and fetal programming. J. Steroid Biochem. Mol. Biol. 2014, 139, 138–143. [Google Scholar] [CrossRef] [PubMed]
  117. Gollenberg, A.L.; Addo, O.Y.; Zhang, Z.; Hediger, M.L.; Himes, J.H.; Lee, P.A. In utero exposure to cigarette smoking, environmental tobacco smoke and reproductive hormones in US girls approaching puberty. Horm. Res. Paediatr. 2015, 83, 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Ernst, A.; Kristensen, S.L.; Toft, G.; Thulstrup, A.M.; Hakonsen, L.B.; Olsen, S.F.; Ramlau-Hansen, C.H. Maternal smoking during pregnancy and reproductive health of daughters: A follow-up study spanning two decades. Hum. Reprod. 2012, 27, 3593–3600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Ittermann, T.; Thamm, M.; Schipf, S.; John, U.; Rettig, R.; Volzke, H. Relationship of smoking and/or passive exposure to tobacco smoke on the association between serum thyrotropin and body mass index in large groups of adolescents and children. Thyroid 2013, 23, 262–268. [Google Scholar] [CrossRef]
  120. Filis, P.; Hombach-Klonisch, S.; Ayotte, P.; Nagrath, N.; Soffientini, U.; Klonisch, T.; O’Shaughnessy, P.; Fowler, P.A. Maternal smoking and high BMI disrupt thyroid gland development. BMC Med. 2018, 16, 194. [Google Scholar] [CrossRef] [Green Version]
  121. Singhal, A.; Farooqi, I.S.; Cole, T.J.; O’Rahilly, S.; Fewtrell, M.; Kattenhorn, M.; Lucas, A.; Deanfield, J. Influence of leptin on arterial distensibility: A novel link between obesity and cardiovascular disease? Circulation 2002, 106, 1919–1924. [Google Scholar] [CrossRef] [Green Version]
  122. Wilce, M.C.; Parker, M.W. Structure and function of glutathione S-transferases. Biochim. Biophys. Acta 1994, 1205, 1–18. [Google Scholar] [CrossRef]
  123. Panasevich, S.; Lindgren, C.; Kere, J.; Wickman, M.; Pershagen, G.; Nyberg, F.; Melen, E. Interaction between early maternal smoking and variants in TNF and GSTP1 in childhood wheezing. Clin. Exp. Allergy 2010, 40, 458–467. [Google Scholar] [CrossRef] [Green Version]
  124. Lee, Y.L.; Lee, Y.C.; Guo, Y.L. Associations of glutathione S-transferase P1, M1, and environmental tobacco smoke with wheezing illness in school children. Allergy 2007, 62, 641–647. [Google Scholar] [CrossRef]
  125. Palmer, C.N.; Doney, A.S.; Lee, S.P.; Murrie, I.; Ismail, T.; Macgregor, D.F.; Mukhopadhyay, S. Glutathione S-transferase M1 and P1 genotype, passive smoking, and peak expiratory flow in asthma. Pediatrics 2006, 118, 710–716. [Google Scholar] [CrossRef]
  126. Wu, C.C.; Ou, C.Y.; Chang, J.C.; Hsu, T.Y.; Kuo, H.C.; Liu, C.A.; Wang, C.L.; Chuang, C.J.; Chuang, H.; Liang, H.M.; et al. Gender-dependent effect of GSTM1 genotype on childhood asthma associated with prenatal tobacco smoke exposure. Biomed. Res. Int. 2014, 2014, 769452. [Google Scholar] [CrossRef] [PubMed]
  127. Lee, S.Y.; Kim, B.S.; Kwon, S.O.; Oh, S.Y.; Shin, H.L.; Jung, Y.H.; Lee, E.; Yang, S.I.; Kim, H.Y.; Seo, J.H.; et al. Modification of additive effect between vitamins and ETS on childhood asthma risk according to GSTP1 polymorphism: A cross-sectional study. BMC Pulm. Med. 2015, 15, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Wenten, M.; Li, Y.F.; Lin, P.C.; Gauderman, W.J.; Berhane, K.; Avol, E.; Gilliland, F.D. In utero smoke exposure, glutathione S-transferase P1 haplotypes, and respiratory illness-related absence among schoolchildren. Pediatrics 2009, 123, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
  129. Turner, S.; Francis, B.; Wani, N.; Vijverberg, S.; Pino-Yanes, M.; Mukhopadhyay, S.; Tavendale, R.; Palmer, C.; Burchard, E.G.; Merid, S.K.; et al. Variants in genes coding for glutathione S-transferases and asthma outcomes in children. Pharmacogenomics 2018, 19, 707–713. [Google Scholar] [CrossRef] [PubMed]
  130. Morales, E.; Sunyer, J.; Julvez, J.; Castro-Giner, F.; Estivill, X.; Torrent, M.; De Cid, R. GSTM1 polymorphisms modify the effect of maternal smoking during pregnancy on cognitive functioning in preschoolers. Int. J. Epidemiol. 2009, 38, 690–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Filonzi, L.; Magnani, C.; Lavezzi, A.M.; Vaghi, M.; Nosetti, L.; Nonnis Marzano, F. Detoxification genes polymorphisms in SIDS exposed to tobacco smoke. Gene 2018, 648, 1–4. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, I.J.; Guo, Y.L.; Lin, T.J.; Chen, P.C.; Wu, Y.N. GSTM1, GSTP1, prenatal smoke exposure, and atopic dermatitis. Ann. Allerg Asthma Immunol. 2010, 105, 124–129. [Google Scholar] [CrossRef]
  133. Li, X.; Liu, Z.; Deng, Y.; Li, S.; Mu, D.; Tian, X.; Lin, Y.; Yang, J.; Li, J.; Li, N.; et al. Modification of the association between maternal smoke exposure and congenital heart defects by polymorphisms in glutathione S-transferase genes. Sci. Rep. 2015, 5, 14915. [Google Scholar] [CrossRef]
  134. Dai, X.; Dharmage, S.C.; Bowatte, G.; Waidyatillake, N.T.; Perret, J.L.; Hui, J.; Erbas, B.; Abramson, M.J.; Lowe, A.J.; Burgess, J.A.; et al. Interaction of Glutathione S-Transferase M1, T1, and P1 Genes With Early Life Tobacco Smoke Exposure on Lung Function in Adolescents. Chest 2019, 155, 94–102. [Google Scholar] [CrossRef]
  135. Wenten, M.; Berhane, K.; Rappaport, E.B.; Avol, E.; Tsai, W.W.; Gauderman, W.J.; McConnell, R.; Dubeau, L.; Gilliland, F.D. TNF-308 modifies the effect of second-hand smoke on respiratory illness-related school absences. Am. J. Respir. Crit. Care Med. 2005, 172, 1563–1568. [Google Scholar] [CrossRef] [Green Version]
  136. Salam, M.T.; Gauderman, W.J.; McConnell, R.; Lin, P.C.; Gilliland, F.D. Transforming growth factor- 1 C-509T polymorphism, oxidant stress, and early-onset childhood asthma. Am. J. Respir. Crit. Care Med. 2007, 176, 1192–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Smith, A.M.; Bernstein, D.I.; LeMasters, G.K.; Huey, N.L.; Ericksen, M.; Villareal, M.; Lockey, J.; Khurana Hershey, G.K. Environmental tobacco smoke and interleukin 4 polymorphism (C-589T) gene: Environment interaction increases risk of wheezing in African-American infants. J. Pediatr. 2008, 152, 709–715. [Google Scholar] [CrossRef] [PubMed]
  138. Sadeghnejad, A.; Karmaus, W.; Arshad, S.H.; Kurukulaaratchy, R.; Huebner, M.; Ewart, S. IL13 gene polymorphisms modify the effect of exposure to tobacco smoke on persistent wheeze and asthma in childhood, a longitudinal study. Respir. Res. 2008, 9, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Wright, S.D.; Ramos, R.A.; Tobias, P.S.; Ulevitch, R.J.; Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef]
  140. Bottema, R.W.; Reijmerink, N.E.; Kerkhof, M.; Koppelman, G.H.; Stelma, F.F.; Gerritsen, J.; Thijs, C.; Brunekreef, B.; van Schayck, C.P.; Postma, D.S. Interleukin 13, CD14, pet and tobacco smoke influence atopy in three Dutch cohorts: The allergenic study. Eur. Respir. J. 2008, 32, 593–602. [Google Scholar] [CrossRef]
  141. Hussein, Y.M.; Shalaby, S.M.; Zidan, H.E.; Sabbah, N.A.; Karam, N.A.; Alzahrani, S.S. CD14 tobacco gene-environment interaction in atopic children. Cell. Immunol. 2013, 285, 31–37. [Google Scholar] [CrossRef]
  142. Bouzigon, E.; Corda, E.; Aschard, H.; Dizier, M.H.; Boland, A.; Bousquet, J.; Chateigner, N.; Gormand, F.; Just, J.; Le Moual, N.; et al. Effect of 17q21 variants and smoking exposure in early-onset asthma. N. Engl. J. Med. 2008, 359, 1985–1994. [Google Scholar] [CrossRef] [Green Version]
  143. Flory, J.H.; Sleiman, P.M.; Christie, J.D.; Annaiah, K.; Bradfield, J.; Kim, C.E.; Glessner, J.; Imielinski, M.; Li, H.; Frackelton, E.C.; et al. 17q12-21 variants interact with smoke exposure as a risk factor for pediatric asthma but are equally associated with early-onset versus late-onset asthma in North Americans of European ancestry. J. Allergy Clin. Immunol. 2009, 124, 605–607. [Google Scholar] [CrossRef]
  144. van der Valk, R.J.; Duijts, L.; Kerkhof, M.; Willemsen, S.P.; Hofman, A.; Moll, H.A.; Smit, H.A.; Brunekreef, B.; Postma, D.S.; Jaddoe, V.W.; et al. Interaction of a 17q12 variant with both fetal and infant smoke exposure in the development of childhood asthma-like symptoms. Allergy 2012, 67, 767–774. [Google Scholar] [CrossRef]
  145. Kreiner-Moller, E.; Strachan, D.P.; Linneberg, A.; Husemoen, L.L.; Bisgaard, H.; Bonnelykke, K. 17q21 gene variation is not associated with asthma in adulthood. Allergy 2015, 70, 107–114. [Google Scholar] [CrossRef]
  146. Morita, H.; Nagai, R. Smoking exposure, 17q21 variants, and early-onset asthma. N. Engl. J. Med. 2009, 360, 1255. [Google Scholar] [CrossRef] [PubMed]
  147. Dizier, M.H.; Bouzigon, E.; Guilloud-Bataille, M.; Siroux, V.; Lemainque, A.; Boland, A.; Lathrop, M.; Demenais, F. Evidence for gene x smoking exposure interactions in a genome-wide linkage screen of asthma and bronchial hyper-responsiveness in EGEA families. Eur. J. Hum. Genet. 2007, 15, 810–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Dizier, M.H.; Nadif, R.; Margaritte-Jeannin, P.; Barton, S.J.; Sarnowski, C.; Gagne-Ouellet, V.; Brossard, M.; Lavielle, N.; Just, J.; Lathrop, M.; et al. Interaction between the DNAH9 gene and early smoke exposure in bronchial hyperresponsiveness. Eur. Respir. J. 2016, 47, 1072–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Dizier, M.H.; Margaritte-Jeannin, P.; Pain, L.; Sarnowski, C.; Brossard, M.; Mohamdi, H.; Lavielle, N.; Babron, M.C.; Just, J.; Lathrop, M.; et al. Interactive effect between ATPase-related genes and early-life tobacco smoke exposure on bronchial hyper-responsiveness detected in asthma-ascertained families. Thorax 2019, 74, 254–260. [Google Scholar] [CrossRef] [PubMed]
  150. Olivo-Marston, S.E.; Yang, P.; Mechanic, L.E.; Bowman, E.D.; Pine, S.R.; Loffredo, C.A.; Alberg, A.J.; Caporaso, N.; Shields, P.G.; Chanock, S.; et al. Childhood exposure to secondhand smoke and functional mannose binding lectin polymorphisms are associated with increased lung cancer risk. Cancer Epidemiol. Prev. Biomark. 2009, 18, 3375–3383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Poetsch, M.; Czerwinski, M.; Wingenfeld, L.; Vennemann, M.; Bajanowski, T. A common FMO3 polymorphism may amplify the effect of nicotine exposure in sudden infant death syndrome (SIDS). Int. J. Legal. Med. 2010, 124, 301–306. [Google Scholar] [CrossRef]
  152. Boneva, R.S.; Botto, L.D.; Moore, C.A.; Yang, Q.; Correa, A.; Erickson, J.D. Mortality associated with congenital heart defects in the United States: Trends and racial disparities, 1979–1997. Circulation 2001, 103, 2376–2381. [Google Scholar] [CrossRef] [Green Version]
  153. Tang, X.; Hobbs, C.A.; Cleves, M.A.; Erickson, S.W.; MacLeod, S.L.; Malik, S.; the National Birth Defects Prevention Study. Genetic variation affects congenital heart defect susceptibility in offspring exposed to maternal tobacco use. Birth Defects Res. Part A Clin. Mol. Teratol. 2015, 103, 834–842. [Google Scholar] [CrossRef] [Green Version]
  154. Satokata, I.; Maas, R. Msx1 deficient mice exhibit cleft palate and abnormalities of craniofacial and tooth development. Nat. Genet. 1994, 6, 348–356. [Google Scholar] [CrossRef]
  155. van den Boogaard, M.J.; de Costa, D.; Krapels, I.P.; Liu, F.; van Duijn, C.; Sinke, R.J.; Lindhout, D.; Steegers-Theunissen, R.P. The MSX1 allele 4 homozygous child exposed to smoking at periconception is most sensitive in developing nonsyndromic orofacial clefts. Hum. Genet. 2008, 124, 525–534. [Google Scholar] [CrossRef] [Green Version]
  156. NCBI. National Center for Biotechnology Information, U.S. National Library of Medicine, Gene. Available online: https://www.ncbi.nlm.nih.gov/gene/ (accessed on 21 February 2020).
  157. O’Shaughnessy, P.J.; Monteiro, A.; Bhattacharya, S.; Fowler, P.A. Maternal smoking and fetal sex significantly affect metabolic enzyme expression in the human fetal liver. J. Clin. Endocrinol. Metab. 2011, 96, 2851–2860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Drake, A.J.; O’Shaughnessy, P.J.; Bhattacharya, S.; Monteiro, A.; Kerrigan, D.; Goetz, S.; Raab, A.; Rhind, S.M.; Sinclair, K.D.; Meharg, A.A.; et al. In utero exposure to cigarette chemicals induces sex-specific disruption of one-carbon metabolism and DNA methylation in the human fetal liver. BMC Med. 2015, 13, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Filis, P.; Nagrath, N.; Fraser, M.; Hay, D.C.; Iredale, J.P.; O’Shaughnessy, P.; Fowler, P.A. Maternal Smoking Dysregulates Protein Expression in Second Trimester Human Fetal Livers in a Sex-Specific Manner. J. Clin. Endocrinol. Metab. 2015, 100, E861–E870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Du, T.; Zamore, P.D. Beginning to understand microRNA function. Cell Res. 2007, 17, 661–663. [Google Scholar] [CrossRef] [PubMed]
  161. Maccani, M.A.; Avissar-Whiting, M.; Banister, C.E.; McGonnigal, B.; Padbury, J.F.; Marsit, C.J. Maternal cigarette smoking during pregnancy is associated with downregulation of miR-16, miR-21, and miR-146a in the placenta. Epigenetics 2010, 5, 583–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Gao, L.; Liu, X.; Millstein, J.; Siegmund, K.D.; Dubeau, L.; Maguire, R.L.; Jim Zhang, J.; Fuemmeler, B.F.; Kollins, S.H.; Hoyo, C.; et al. Self-reported prenatal tobacco smoke exposure, AXL gene-body methylation, and childhood asthma phenotypes. Clin. Epigenet. 2018, 10, 98. [Google Scholar] [CrossRef] [Green Version]
  163. Mudduluru, G.; Ceppi, P.; Kumarswamy, R.; Scagliotti, G.V.; Papotti, M.; Allgayer, H. Regulation of Axl receptor tyrosine kinase expression by miR-34a and miR-199a/b in solid cancer. Oncogene 2011, 30, 2888–2899. [Google Scholar] [CrossRef]
  164. Liu, Q.; Wang, W.; Jing, W. Indoor air pollution aggravates asthma in Chinese children and induces the changes in serum level of miR-155. Int. J. Environ. Health Res. 2019, 29, 22–30. [Google Scholar] [CrossRef]
  165. Jylhava, J.; Pedersen, N.L.; Hagg, S. Biological Age Predictors. EBioMedicine 2017, 21, 29–36. [Google Scholar] [CrossRef] [Green Version]
  166. Osorio-Yanez, C.; Clemente, D.B.P.; Maitre, L.; Vives-Usano, M.; Bustamante, M.; Martinez, D.; Casas, M.; Alexander, J.; Thomsen, C.; Chatzi, L.; et al. Early life tobacco exposure and children’s telomere length: The HELIX project. Sci. Total Environ. 2020, 711, 135028. [Google Scholar] [CrossRef]
  167. Breitling, L.P.; Yang, R.; Korn, B.; Burwinkel, B.; Brenner, H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am. J. Hum. Genet. 2011, 88, 450–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Martino, D.; Prescott, S. Epigenetics and prenatal influences on asthma and allergic airways disease. Chest 2011, 139, 640–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Ganu, R.S.; Harris, R.A.; Collins, K.; Aagaard, K.M. Early origins of adult disease: Approaches for investigating the programmable epigenome in humans, nonhuman primates, and rodents. ILAR J. 2012, 53, 306–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Wadhwa, P.D.; Buss, C.; Entringer, S.; Swanson, J.M. Developmental origins of health and disease: Brief history of the approach and current focus on epigenetic mechanisms. Semin. Reprod. Med. 2009, 27, 358–368. [Google Scholar] [CrossRef] [Green Version]
  171. Nielsen, C.H.; Larsen, A.; Nielsen, A.L. DNA methylation alterations in response to prenatal exposure of maternal cigarette smoking: A persistent epigenetic impact on health from maternal lifestyle? Arch. Toxicol. 2016, 90, 231–245. [Google Scholar] [CrossRef]
  172. Breton, C.V.; Byun, H.M.; Wenten, M.; Pan, F.; Yang, A.; Gilliland, F.D. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am. J. Respir. Crit. Care Med. 2009, 180, 462–467. [Google Scholar] [CrossRef]
  173. Suter, M.; Abramovici, A.; Showalter, L.; Hu, M.; Shope, C.D.; Varner, M.; Aagaard-Tillery, K. In utero tobacco exposure epigenetically modifies placental CYP1A1 expression. Metabolism 2010, 59, 1481–1490. [Google Scholar] [CrossRef] [Green Version]
  174. Suter, M.; Ma, J.; Harris, A.; Patterson, L.; Brown, K.A.; Shope, C.; Showalter, L.; Abramovici, A.; Aagaard-Tillery, K.M. Maternal tobacco use modestly alters correlated epigenome-wide placental DNA methylation and gene expression. Epigenetics 2011, 6, 1284–1294. [Google Scholar] [CrossRef] [Green Version]
  175. Murphy, S.K.; Adigun, A.; Huang, Z.Q.; Overcash, F.; Wang, F.; Jirtle, R.L.; Schildkraut, J.M.; Murtha, A.P.; Iversen, E.S.; Hoyo, C. Gender-specific methylation differences in relation to prenatal exposure to cigarette smoke. Gene 2012, 494, 36–43. [Google Scholar] [CrossRef] [Green Version]
  176. Wang, I.J.; Chen, S.L.; Lu, T.P.; Chuang, E.Y.; Chen, P.C. Prenatal smoke exposure, DNA methylation, and childhood atopic dermatitis. Clin. Exp. Allergy 2013, 43, 535–543. [Google Scholar] [CrossRef]
  177. Novakovic, B.; Ryan, J.; Pereira, N.; Boughton, B.; Craig, J.M.; Saffery, R. Postnatal stability, tissue, and time specific effects of AHRR methylation change in response to maternal smoking in pregnancy. Epigenetics 2014, 9, 377–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Joubert, B.R.; Haberg, S.E.; Nilsen, R.M.; Wang, X.; Vollset, S.E.; Murphy, S.K.; Huang, Z.; Hoyo, C.; Midttun, O.; Cupul-Uicab, L.A.; et al. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ. Health Perspect. 2012, 120, 1425–1431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Markunas, C.A.; Xu, Z.; Harlid, S.; Wade, P.A.; Lie, R.T.; Taylor, J.A.; Wilcox, A.J. Identification of DNA methylation changes in newborns related to maternal smoking during pregnancy. Environ. Health Perspect. 2014, 122, 1147–1153. [Google Scholar] [CrossRef] [PubMed]
  180. Zakarya, R.; Adcock, I.; Oliver, B.G. Epigenetic impacts of maternal tobacco and e-vapour exposure on the offspring lung. Clin. Epigenet. 2019, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  181. Den Dekker, H.T.; Burrows, K.; Felix, J.F.; Salas, L.A.; Nedeljkovic, I.; Yao, J.; Rifas-Shiman, S.L.; Ruiz-Arenas, C.; Amin, N.; Bustamante, M.; et al. Newborn DNA-methylation, childhood lung function, and the risks of asthma and COPD across the life course. Eur. Respir. J. 2019, 53. [Google Scholar] [CrossRef] [Green Version]
  182. Runyon, R.S.; Cachola, L.M.; Rajeshuni, N.; Hunter, T.; Garcia, M.; Ahn, R.; Lurmann, F.; Krasnow, R.; Jack, L.M.; Miller, R.L.; et al. Asthma discordance in twins is linked to epigenetic modifications of T cells. PLoS ONE 2012, 7, e48796. [Google Scholar] [CrossRef] [Green Version]
  183. Reinius, L.E.; Gref, A.; Saaf, A.; Acevedo, N.; Joerink, M.; Kupczyk, M.; D’Amato, M.; Bergstrom, A.; Melen, E.; Scheynius, A.; et al. DNA methylation in the Neuropeptide S Receptor 1 (NPSR1) promoter in relation to asthma and environmental factors. PLoS ONE 2013, 8, e53877. [Google Scholar] [CrossRef]
  184. Haghighi, A.; Melka, M.G.; Bernard, M.; Abrahamowicz, M.; Leonard, G.T.; Richer, L.; Perron, M.; Veillette, S.; Xu, C.J.; Greenwood, C.M.; et al. Opioid receptor mu 1 gene, fat intake and obesity in adolescence. Mol. Psychiatry 2014, 19, 63–68. [Google Scholar] [CrossRef] [Green Version]
  185. Haghighi, A.; Schwartz, D.H.; Abrahamowicz, M.; Leonard, G.T.; Perron, M.; Richer, L.; Veillette, S.; Gaudet, D.; Paus, T.; Pausova, Z. Prenatal exposure to maternal cigarette smoking, amygdala volume, and fat intake in adolescence. JAMA Psychiatry 2013, 70, 98–105. [Google Scholar] [CrossRef] [Green Version]
  186. Lee, K.W.; Abrahamowicz, M.; Leonard, G.T.; Richer, L.; Perron, M.; Veillette, S.; Reischl, E.; Bouchard, L.; Gaudet, D.; Paus, T.; et al. Prenatal exposure to cigarette smoke interacts with OPRM1 to modulate dietary preference for fat. J. Psychiatry Neurosci. 2015, 40, 38–45. [Google Scholar] [CrossRef] [Green Version]
  187. Oken, E.; Levitan, E.B.; Gillman, M.W. Maternal smoking during pregnancy and child overweight: Systematic review and meta-analysis. Int. J. Obes. 2008, 32, 201–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. COPHES. Consortium to Perform Human Biomonitoring on a European Scale. 2012. Available online: http://eu-hbm.info/cophes (accessed on 16 April 2020).
  189. Joas, R.; Casteleyn, L.; Biot, P.; Kolossa-Gehring, M.; Castano, A.; Angerer, J.; Schoeters, G.; Sepai, O.; Knudsen, L.E.; Joas, A.; et al. Harmonised human biomonitoring in Europe: Activities towards an EU HBM framework. Int. J. Hyg. Environ. Health 2012, 215, 172–175. [Google Scholar] [CrossRef] [PubMed]
  190. DiMoPEx. Diagnosis, Monitoring and Prevention of Exposure Related Non-Communicable Diseases, DiMoPEx (CA 15129). 2020. Available online: http://dimopex.eu/about/ (accessed on 16 April 2020).
  191. Budnik, L.T.; Adam, B.; Albin, M.; Banelli, B.; Baur, X.; Belpoggi, F.; Bolognesi, C.; Broberg, K.; Gustavsson, P.; Goen, T.; et al. Diagnosis, monitoring and prevention of exposure-related non-communicable diseases in the living and working environment: DiMoPEx-project is designed to determine the impacts of environmental exposure on human health. J. Occup. Med. Toxicol. 2018, 13, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Adverse health effects in children induced by second-hand smoke and prenatal tobacco smoke exposure. COPD = chronic obstructive pulmonary disease.
Figure 1. Adverse health effects in children induced by second-hand smoke and prenatal tobacco smoke exposure. COPD = chronic obstructive pulmonary disease.
Ijerph 17 03212 g001
Table 1. Summary of changes in children induced by second-hand smoke (SHS) and prenatal tobacco smoke (PTS) exposure.
Table 1. Summary of changes in children induced by second-hand smoke (SHS) and prenatal tobacco smoke (PTS) exposure.
TopicEffects on ChildrenTest MaterialSourceAssociation
Biomarkers
Matrix metalloproteinase-9 (MMP-9)MMP-9 increasedNasal secretionSHSAllergy, asthma, chronic bronchitis [26], no effect [29]
Cytokine Interleukin (IL)IL-1β increased SalivaSHSInflammatory processes [35,36]
IL-1β decreased BloodSHSInflammatory processes [34]
IL-4 decreased BloodSHSInflammatory processes [34]
IL-5 decreased BloodSHSInflammatory processes [34]
IL-6 increased SalivaSHSInflammatory processes [36]
IL-8 increased New-born dried bloodPTSInflammatory processes [33]
IL-8 Nasal secretionSHSno effect by SHS [29]
IL-8 SalivaSHSno effect by SHS [36]
IL-13 increased Airway secretionSHSInflammatory processes [37]
IL-17 Nasal secretionSHSno effect by SHS [29]
Cytokine Interferon gamma (IFN-γ)IFN-γ decreased BloodSHSInflammatory processes [34]
Tumour necrosis factor alpha (TNF-α)TNF-α increased SalivaSHSInflammatory processes [36]
Urinary leukotriene E4 (uLTE4)uLTE4 increased UrineSHSAsthma [42,43]
Estimated glomerular filtration rate (eGFR)eGFR decreased Serum SHSKidney function, proteinuria [55,56]
Intercellular adhesion molecule 1 (s-ICAM1)s-ICAM1 increased SerumSHSEndothelial stress [61]
Intima-media thickness (IMT)IMT increased UltrasonographySHS, PTSAtherosclerosis [64,65]
C-reactive protein (CRP)CRP increased SerumSHSInflammation response [69,71,72]
Immune status
Regulatory T-cells (Tregs)Treg cell number decreased Cord blood, bloodPTS, SHSAtopy, asthma [74,75,78]
T-helper 17 (Th17) cells Th17 cell number increasedBloodSHSAsthma severity [78]
T-cells subsetsCirculating CD3+ and CD4+ memory T-cell number decreased BloodSHSSystemic immunological response [80]
Circulating CD3+ and CD4+ naïve T-cell number increased BloodSHSSystemic immunological response [80]
CD4+CD45RA+ T-cell number increased BloodSHSSystemic immunological response [80]
CD8+ T-cell number decreased AdenoidsSHSSystemic immunological response [81]
Immunoglobulins A and M (IgA, IgM)IgA and IgM increased AdenoidsSHSSystemic immunological response [82]
Immunoglobulin E (IgE)Immune response to allergens increased SerumSHSAllergy [83,84,85]
Lipid profile
High-density lipoprotein-cholesterol (HDL-C)HDL-C decreasedBloodSHS, PTSArteriosclerosis, obesity, metabolic syndrome [86,87,88,89,90,91,93], no effect by SHS [95]
Low-density lipoprotein-cholesterol (LDL-C)LDL-C increased BloodSHS, PTSArteriosclerosis, obesity, metabolic syndrome [87], no effect by SHS [95]
TriglyceridesTriglycerides increased BloodSHS, PTSArteriosclerosis, obesity, metabolic syndrome [87,88,93], no effect by SHS [95]
Apolipoprotein A-1 (ApoA-1)ApoA-1 decreased BloodSHSArteriosclerosis, obesity, metabolic syndrome [70]
Apolipoprotein B (ApoB)ApoB increased BloodSHSArteriosclerosis, obesity, metabolic syndrome [92]
Oxidative stress (OS) increased In general: cell, tissue and organ injury, cell death; asthma, COPD, cardiovascular events, metabolic syndrome [100,101,102,107]
Nicotinamide adenine dinucleotide phosphate oxidase-2 (Nox2)Nox2 increased SerumSHSArtery dilation [104]
AdiponectinAdiponectin decreased Cord bloodPTSLipid peroxidation increased [108]
Pre-B-cell colony enhancing factor (Visfatin)Visfatin increased Cord bloodPTSLipid peroxidation increased [108]
Urinary 15-F2t-isoprostaneUrinary 15-F2t-isoprostane increased UrineSHSLower lung function parameters [109]
Histone deacetylase-2 (HDAC2)HDAC2 decreased Broncho-alveolar lavage fluidSHSCorticosteroid-insensitiveness leads to impairment of severe asthma treatment [110]
Hormonal changes In general: Metabolic and endocrine dysfunction (foetal, in childhood, and later life) [114,116]
GhrelinGhrelin increased until early adulthood by PTS exposure PlasmaPTSMetabolic disorders [115]
LeptinLeptin increased PlasmaSHSImpairing of vascular function, BMI [70]
AdiponectinAdiponectin decreased Cord bloodPTSOS increased, lipid peroxidation increased [108]
Luteinizing hormone (LH)In girls, LH decreased by PTS exposure but increased by current SHS exposure BloodPTS, SHSReproductive development [117]
Inhibin B (InB)In girls, InB decreased by PTS exposure with no effect by current SHS exposure BloodPTS, SHSReproductive development [117]
Thyrotropin (TSH)TSH decreased SerumSHSHypothyroidism, BMI [119]
Foetal triiodothyronine (T3), thyroxine (T4) and TSHT3, T4 and TSH decreased (possibly by downregulation of foetal thyroid transcripts GATA6 and NKX2-1) Foetal plasmaPTSDisorder of foetal thyroid development and endocrine function [120]
Foetal corticotropin-releasing hormone (CRH), adrenocorticotrophin (ACTH), cortisol, gonadotropins, androgens, oestrogensChanges in foetal steroidogenesis Foetal plasmaPTSMultiple pathophysiological effects (foetal and later in life) by endocrine dysfunction [116]
Table 2. Summary of addressed single nucleotide polymorphisms (SNPs) regarding genetic predisposition to tobacco smoke susceptibility.
Table 2. Summary of addressed single nucleotide polymorphisms (SNPs) regarding genetic predisposition to tobacco smoke susceptibility.
Gene (Chromosome)SNPRisk AlleleAssociation
GSTP1 Exon 5 (11q13)rs1695 (Val-105 or Ile105Val)AG (Ile105Val)Early childhood wheezing [123]; protection against respiratory illness was lost by PTS exposure [128]; no effect in asthma [129]
GG (Val105Val)Asthma [125]; no effect in asthma [129]; current and ever wheezing [124]
AA (Ile105Ile)+ low vitamin A intake: asthma [127]; no effect in asthma [129]; current wheezing [124]; atopic dermatitis [132]; lung function impairment in later life [134]
GSTP1 Intron 5 (11q13)rs749174TTEarly childhood wheezing [123]
GSTP1 Intron 6 (11q13)rs1871042TTEarly childhood wheezing [123]
TNF Promoter (6p21)rs1800629 (-308)AA/AGRespiratory illness [135]
TNF Promoter (6p21)rs1799724 (T-857C)CCEarly childhood wheezing [123]
TNF Intron 1 (6p21)rs1800610CCEarly childhood wheezing [123]
TNF Intron 3 (6p21)rs3093664AG/GGEarly childhood wheezing [123]
TGFB1 Promoter (19q13)rs4803457 (C-509T)TTAsthma [136]
IL-4 (5q31)rs2243250 (C-589T)TT/CTWheezing [137]
IL-13 Exon 4 (5q31)rs20541 (G/A)GGEarly onset persistent wheeze and persistent asthma [138]
IL-13 haplotype pair (Promoter, Intron 1, Exon 4) (5q31)rs1800925 (C/T), rs2066960 (C/A), rs20541 (G/A)CCG/CCGEarly onset persistent wheeze and persistent asthma [138]
CD14 (5q31)3’untranslated region (UTR)AALower IgE levels [140]
CD14 Promoter (5q31)rs2569190 (C-159T)TTElevated IgE levels, atopy [141]
CD14 (5q31)C-550TTTElevated IgE levels, atopy [141]
IKZF3 Intron 3 (17q21)rs9303277CIncreased risk of early-onset asthma enhanced by SHS [142]; confirmed in Caucasians without age of onset [143]
ZPBP2 Exon 2 (17q21)rs11557467 (I151S)GIncreased risk of early-onset asthma enhanced by SHS [142]; confirmed in Caucasians without age of onset [143]
GSDMB Exon 8 (17q21)rs2305480 (P298S)GIncreased risk of early-onset asthma enhanced by SHS [142]; confirmed in Caucasians without age of onset [143]; asthma-like symptoms [144]
GSDMB Exon 8 (17q21)rs2305479 (G291R)CIncreased risk of early-onset asthma enhanced by SHS [142]
GSDMB Intron (17q21)rs4795400CIncreased risk of early-onset asthma enhanced by SHS [142]
GSDMB Intron (17q21)rs9303281AIncreased risk of early-onset asthma enhanced by SHS [142]
GSDMB Intron 1 (17q21)rs7219923TIncreased risk of early-onset asthma enhanced by SHS [142]
GSDMB Intron 2 (17q21)rs2290400CIncreased risk of asthma in Caucasians enhanced by SHS [143]
GSDMB Intron 2 (17q21)rs7216389TIncreased risk of asthma in Caucasians enhanced by SHS [143,145]
GSDMA Exon 2 (17q21)rs3894194AIncreased risk of asthma in Caucasians enhanced by SHS [143]
GSDMA Intron 6 (17q21)rs3859192?Increased risk of asthma in Caucasians enhanced by SHS [143]
ORMDL3 Intron (17q21)rs8076131AIncreased risk of early-onset asthma enhanced by SHS [142]
LRRC3C Intron (17q21)rs8079416?Increased risk of asthma in Caucasians enhanced by SHS [143]
Intergenic region (17q21)rs8069176GIncreased risk of early-onset asthma enhanced by SHS [142]
Intergenic region (17q21)rs4795405CIncreased risk of early-onset asthma enhanced by SHS [142]; confirmed in Caucasians without age of onset [143]
Intergenic region (17q21)rs4794820GIncreased risk of early-onset asthma enhanced by SHS [142]
Intergenic region (17q21)rs8067378?Increased risk of asthma in Caucasians enhanced by SHS [143]
DNAH9 Intron (17p11)rs7225157?Bronchial hyperresponsiveness [148]
ATP8A1 Intron (4p13)rs17448506?Bronchial hyperresponsiveness [149]
ABCA1 Intron (9q31)rs2253304?Bronchial hyperresponsiveness [149]
MBL2 (10q21)rs5030737AAIncreased risk of lung cancer in later life [150]
MBL2 Intron (10q21)rs1838066CCIncreased risk of lung cancer in later life [150]
MBL2 Intron (10q21)rs7095891TTIncreased risk of lung cancer in later life [150]
MBL2 (10q21)rs2165810TTIncreased risk of lung cancer in later life [150]
FMO3 (1q24)rs2266782 (G472A)AARisk factor for sudden infant death syndrome [151]
OSGEP Intron (14q11)rs1320150AGIncreased risk of congenital heart defects [153]
OSGEP Intron (14q11)rs938881?Increased risk of congenital heart defects [153]
OSGEP (14q11)rs2275007?Increased risk of congenital heart defects [153]
OSGEP Intron (14q11)rs883037?Increased risk of congenital heart defects [153]
MSX1 Intron allele 4 (4p16) Homozygosity of 9 repeats of the A4 CA marker Increased risk of nonsyndromic orofacial clefts [155]
Additional information was given from the National Center for Biotechnology Information (NCBI) of the U.S. National Library of Medicine database [156]. The table presents risk alleles associated with a disease. ? = risk allele not reported. SNP = single nucleotide polymorphism. A = adenine. C = cytosine. G = guanine. T = thymine.

Share and Cite

MDPI and ACS Style

Braun, M.; Klingelhöfer, D.; Oremek, G.M.; Quarcoo, D.; Groneberg, D.A. Influence of Second-Hand Smoke and Prenatal Tobacco Smoke Exposure on Biomarkers, Genetics and Physiological Processes in Children—An Overview in Research Insights of the Last Few Years. Int. J. Environ. Res. Public Health 2020, 17, 3212. https://doi.org/10.3390/ijerph17093212

AMA Style

Braun M, Klingelhöfer D, Oremek GM, Quarcoo D, Groneberg DA. Influence of Second-Hand Smoke and Prenatal Tobacco Smoke Exposure on Biomarkers, Genetics and Physiological Processes in Children—An Overview in Research Insights of the Last Few Years. International Journal of Environmental Research and Public Health. 2020; 17(9):3212. https://doi.org/10.3390/ijerph17093212

Chicago/Turabian Style

Braun, Markus, Doris Klingelhöfer, Gerhard M. Oremek, David Quarcoo, and David A. Groneberg. 2020. "Influence of Second-Hand Smoke and Prenatal Tobacco Smoke Exposure on Biomarkers, Genetics and Physiological Processes in Children—An Overview in Research Insights of the Last Few Years" International Journal of Environmental Research and Public Health 17, no. 9: 3212. https://doi.org/10.3390/ijerph17093212

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