Next Article in Journal
Repurposing of the Drug Tezosentan for Cancer Therapy
Next Article in Special Issue
Disruption of the Functional Activity of Neutrophil Granulocytes as a Risk Factor for the Development of Lung Damage in Pregnant Women with COVID-19
Previous Article in Journal
Potential Diagnostic Value of Salivary Tumor Markers in Breast, Lung and Ovarian Cancer: A Preliminary Study
Previous Article in Special Issue
Abietane Diterpenoids Isolated from Torreya nucifera Disrupt Replication of Influenza Virus by Blocking the Phosphatidylinositol-3-Kinase (PI3K)-Akt and ERK Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antioxidant Intake and Biomarkers of Asthma in Relation to Smoking Status—A Review

by
Naser A. Alsharairi
Heart, Mind & Body Research Group, Griffith University, Gold Coast P.O. Box 4222, QLD, Australia
Curr. Issues Mol. Biol. 2023, 45(6), 5099-5117; https://doi.org/10.3390/cimb45060324
Submission received: 26 May 2023 / Revised: 7 June 2023 / Accepted: 9 June 2023 / Published: 10 June 2023

Abstract

:
Asthma is considered a chronic inflammatory disorder associated with airway hyperresponsiveness (AHR). Increased oxidative stress (OS) is a clinical feature of asthma, which promotes the inflammatory responses in bronchial/airway epithelial cells. Smokers and nonsmokers with asthma have been shown to have increases in several OS and inflammatory biomarkers. However, studies suggest significant differences in OS and inflammation biomarkers between smokers and nonsmokers. A few studies suggest associations between antioxidant intake from diet/supplements and asthma in patients with different smoking status. Evidence is lacking on the protective role of antioxidant vitamin and/or mineral consumption against asthma by smoking status with respect to inflammation and OS biomarkers. Therefore, the aim of this review is to highlight current knowledge regarding the relations between antioxidant intake, asthma, and its associated biomarkers, according to smoking status. This paper can be used to guide future research directions towards the health consequences of antioxidant intake in smoking and nonsmoking asthmatics.

1. Introduction

Smoking is regarded as a significant risk factor for asthma progression [1]. The number of asthma deaths due to smoking in 2019 was higher in men than in women [1,2]. Asthma is characterized by airway hyperresponsiveness (AHR) and reversible airflow obstruction, which is attributed to increased airway smooth muscle (ASM) contraction [3,4,5]. Asthma is associated predominantly with mast/CD4+ cells, T lymphocytes and eosinophils. Mucous hypersecretion, luminal obstruction, goblet cell hyperplasia, and thickening of bronchial walls are commonly observed features in asthma [3].
Tobacco smoke is associated with reduced lung function measured as forced expiratory volume in 1 s (FEV1) and increased bronchial hyperresponsiveness in smokers with asthma [6]. Asthmatic patients who smoked ≥10 pack/year had a rapid decline in FEV1 and forced vital capacity (FVC) compared with those who smoked <10 pack/year after 12-year follow-up [7]. Secondhand smoke (SHS) exposure has been linked to asthma risk in active and/or former smokers [8]. Exposure to SHS in public places was associated with a marked decrease in peak expiratory flow rate (PEFR) and FVC in asthmatic smokers [9]. The risk of asthma among nonsmokers who were exposed to SHS has increased in a large adult-onset asthma population with 16 years of follow-up [10].
Tobacco smoke consists of a range of toxic chemicals (e.g., benzopyrene, acrolein, crotonaldehyde, phenols, ammonia, nitrosamines, hydrocarbons, aromatic amines), which are potentially harmful to human bronchial epithelial cells (HBECs), causing airway inflammation by increasing mitochondrial reactive oxygen species (ROS) and pro-inflammatory interleukin (IL)-8 cytokine production [11,12]. Downregulation of microRNAs in lung fibroblasts of smokers may affect its function due to aberrant DNA methylation at specific sites [13]. Moderate asthma was associated with lung inflammation, and this response is related to reduced expression of microRNA target genes such as I-miR-146a [14]. Cigarette smoke extract (CSE) exposure in HBECs results in increased oxidative stress (OS) and pro-inflammatory cytokines IL-6, IL-8, and tumor necrosis factor α (TNF-α) by the activation of several inflammatory signaling pathways, including the transcription factor-kappaB (NF-κB), extracellular signal-regulated kinases (ERK 1/2), c-Jun N-terminal kinase (JNK), and mitogen-activated protein kinases (MAPKs) [15]. Tobacco smoke alters immune responses in the lung, triggering asthma by activating Toll-like receptors (e.g., TLR-2 and TLR-4)-stimulated pro-inflammatory cytokine production and increasing total serum immunoglobulin E (IgE) levels in airway epithelial cells [16]. In asthmatic patients, exposure to environmental tobacco smoke (ETS) results in oxidant/antioxidant imbalance, which leads to increased pro-inflammatory biomarkers as assessed by increased TNFα, IL-6, and IL-8 [16]. Evidence suggests that nicotine is not carcinogenic, but it may affect the airway epithelial cells of asthmatic smokers by activating nitrosamine 4(methylnitrosamino)-1-(3–pyridyl)-1-butanone (NNK), which binds to the α7 nicotinic acetylcholine receptor (α7nAChR), leading to AHR and inflammation by upregulating the α7nAChR-mediated signaling pathways [17].
The genetic variants–tobacco smoke exposure interaction has been shown to increase asthma risk in smokers and nonsmokers. Evidence of the interaction between variants of rs9969775 on chromosome 9, rs5011804 on chromosome 12, and active tobacco smoking was reported in asthmatic adults [18]. Genetic variants of NLR Family CARD Domain Containing 4 (NLRP4) inflammasome are implicated in asthma exacerbation in current and former adult smokers as evidenced by high genotype-specific expression of rs16986718G [19]. The presence of mutant AG/GG genotype for CD14 rs2569190 and rs13150331 (TLR) polymorphism in asthmatic adult smokers increases the risk of the disease [20]. Asthmatic nonsmokers carrying allele homozygotes of rs1384006 C  >  T of the OS responsive kinase 1 (OXSR1) gene are at higher asthma exacerbation risk than asthmatic smokers [21].
Few studies have evaluated evidence-based treatment for asthma in smokers. Pycnogenol®, a herbal dietary supplement-based extract manufactured by Horphag Research (Geneva, Switzerland) and derived from French Pinus pinaster bark, is regarded as an option for the treatment of asthma when used in combination with the inhalation corticosteroid (ICS) therapy, resulting in improvement of asthma symptoms [22]. Asthmatic smokers have ICS insensitivity as compared to asthmatic nonsmokers and are less responsive to the benefits of ICS treatment alone. Alterations of inflammatory phenotypes and glucocorticoid receptors and the reduction of histone deacetylase (HDAC) activity are considered potential mechanisms of corticosteroid insensitivity in asthmatic smokers [23,24]. The combination of ICS therapy and a long-acting β2 adrenergic (LABA) displays a better clinical improvement for smoking and nonsmoking asthmatics than using ICS alone [25,26]. The use of nicotine replacement therapy, varenicline or bupropion, may significantly improve lung function and AHR in asthmatic smokers [25].
There is still a significant amount of uncertainty in the safety and efficacy of dietary supplements for the treatment of lung diseases among smokers and/or nonsmokers due to the limited number randomized controlled trials (RCTs) [27,28]. Thus, there is a need to focus on the role of antioxidants in smoking-related asthma risk. A recent review investigating the effects of dietary antioxidant intake on lung cancer (LC) risk among smokers and nonsmokers suggests that dietary vitamins (C, D, E, and carotenoids) and minerals (zinc and copper) may exert protective effects against cigarette smoke (CS)-induced OS and/or inflammation. However, dietary retinol and iron intake did not provide any protection, and research suggests caution in recommending these for LC treatment [29]. There is a direct association between LC and asthma in smokers [30,31]. Given that smoking is considered a risk factor for asthma through increased levels of OS and inflammatory cytokine production [17], targeting dietary/supplement-derived antioxidants might help our understanding of their role in protecting bronchial epithelial cells against CS-induced-OS/inflammatory biomarkers in smokers and nonsmokers. This paper explores this connection to gain insight into the health consequences of antioxidant consumption and makes recommendations for future studies. To date, there have been no reviews to evaluate antioxidant intake and the biomarkers of OS and inflammation in asthma, according to smoking status.

2. Methods

A literature review in the PubMed/MEDLINE database and Google Scholar was conducted for English language studies published between 1 January 2000 and 30 April 2023. The following search terms were used: asthma, diet, supplements, antioxidant, vitamins, minerals, OS, lipid peroxidation (LP), inflammation, biomarkers, antioxidant/oxidant enzymes, bronchial/airway epithelial cells, smoking, smokers, and nonsmokers. Studies focusing on the chronic obstructive pulmonary disease (COPD) were excluded, as the diagnostic biomarkers in asthma are different from both COPD and the asthma-COPD overlap. All studies relevant to the search terms were included, and the search was not restricted to a particular study design.

3. Antioxidant Intake and Asthma in Relation to Smoking Status

Studies investigating the associations between antioxidant intake and asthma according to smoking status are limited. Smokers with low dietary vitamin C (VC) intake had chronic bronchitis symptoms associated with asthma compared with those who had higher intake [32]. According to quartiles of carotenoid dietary/supplement intake (carotene, lycopene, and lutein with zeaxanthin), the risk of asthma was reported to be lower in the fourth quartile (≥165.59 μg/kg per day) than the first quartile (<41.43 (μg/kg per day) among current smokers, ex-smokers, and nonsmokers with asthma [33]. One trial revealed no effects of 6 weeks of supplemental vitamin E (VE) on AHR in nonsmokers with asthma [34]. Supplementation with selenium (Se) had no significant improvement in asthma-related quality of life (QoL) and lung function regardless of smoking status [35]. These findings suggest that dietary VC and carotenoids intake may reduce asthma in smokers and/or nonsmokers. Supplementation with VE and Se had no effect on asthma in smokers and nonsmokers. The associations between antioxidant intake and asthma risk according to smoking status are summarized in Table 1.

4. Biomarkers of OS and Inflammation in Relation to Smoking Status

OS is regarded as the major contributor to CS-induced airway inflammation [36]. Evidence from many studies, mostly derived from case-control design, has shown that CS activates OS by augmenting airway inflammation in smoking and nonsmoking asthmatics.

4.1. Biomarkers of OS

Case-Control Studies

Asthmatic current smokers showed increased serum levels of malondialdehyde (MDA) and decreased levels of the ferric-reducing ability of plasma (FRAP) [37]. Higher MDA levels in exhaled breath condensate (EBC) have been reported in active smoking asthmatics than in their ex-smoking and nonsmoking counterparts [38]. The levels of protein carbonyls and peroxynitrite in plasma were reported to be higher in current smoking asthmatics than in their ex-smoking and nonsmoking counterparts [39]. Smoking asthmatics with a lower FEV1 have higher erythrocyte antioxidant enzyme activity, including superoxide dismutase (SOD) and disulfide/oxidized glutathione (GSH) activity, than nonsmoking patients with asthma and healthy controls. Increased SOD and GSH in smoking asthmatics may not protect airway epithelial cells against the harmful effects of free radicals. SOD and GSH activities were found to be higher in nonsmoking asthmatics than healthy controls [40]. Nonsmoking asthmatics demonstrated increased levels of nitrite (NO2), protein carbonyls, lipid peroxide, SOD activity, and decreased protein sulfhydrils and glutathione peroxidase (GPx) activity in leukocytes and red blood cells [41]. High sputum GSH and NO2 levels were reported in nonsmokers with stable and acute asthma [42].
Overall findings suggest that the oxidant/antioxidant imbalance derived by CS is likely to exist in smoking and nonsmoking asthmatics. OS biomarkers are increased in current and nonsmokers, but the increase in the enzymatic antioxidants in smokers may be insufficient to protect bronchial/airway epithelial cells against oxidative damage. Table 2 shows the OS biomarkers in smoking and nonsmoking asthmatics.

4.2. Biomarkers of Inflammation

4.2.1. Case-Control Studies

Fractional exhaled nitric oxide (FeNO) was reported in lower levels in smoking asthmatics than nonsmoking asthmatics and healthy controls. This reduction is accompanied by increased numbers of sputum eosinophils [43]. Smoking asthmatics had lower FeNO levels than their nonsmoking counterparts, but this decrease does not appear to reflect improvement of asthma control [44]. Low levels of FeNO were observed in current/ex-smokers with severe asthma compared to nonsmokers with mild-moderate asthma. High FeNO levels and blood eosinophil count provide a moderate prediction of type 2 high status in severe asthmatic nonsmokers [45]. Increased levels of FeNO, eosinophils, and neutrophils have been observed in the airways of current nonsmoking asthmatics [46]. Compared to active smokers with asthma, nonsmokers with asthma had higher FeNO levels [47].
Eotaxin-1 in EBC was associated with blood eosinophil count, FeNO value, and serum ECP in nonsmoking asthmatics [48]. Eotaxin was found at higher levels in the sputum of smokers than nonsmokers with asthma. Sputum and serum IL-5 levels were found to be higher in nonsmoking asthmatics than in smoking asthmatics and healthy controls. High BALF eotaxin-1 was associated with increased BALF eosinophil and neutrophil counts and percentages [49]. Comparatively greater levels of sputum IL-1β, IL-5, and Interleukin 18 receptor 1 (IL-18R1) have been reported in current/ex-smoking severe asthmatics compared to healthy controls. Bronchoscopy and Bronchoalveolar Lavavge Fluid (BALF) levels of eotaxin-1 were observed to be high in nonsmoking asthmatics. Higher sputum levels of IL-4, IL-5, IL-1β, Interleukin 1 receptor-like 1 (IL-1RL1), Interleukin 1 receptor, type I (IL-1R1), IL-1R2, IL-18R1, and NLRP3 were detected in nonsmoking severe asthmatics compared to mild-moderate asthmatics and healthy controls [50].
Higher sputum eosinophils, eosinophilic cationic protein (ECP), neutrophils, and IL-8 levels were observed in asthmatic smokers compared to healthy nonsmokers, which were associated with FEV1 and neutrophil count. Compared to healthy nonsmokers, nonsmoking asthmatics demonstrated higher sputum ECP and eosinophil levels [51]. Serum periostin has been observed in higher levels in nonsmokers than smokers with asthma [52]. High serum periostin, TNFα, IL-4, IL-5, and the chitinase-like protein YKL-40 levels, as well as low serum IL-37 levels, were associated with exacerbated asthma in nonsmokers [53]. Asthmatic smokers demonstrated increased sputum levels of neutrophils, and decreased levels of eosinophils compared to asthmatic nonsmokers. Both asthmatic smokers and nonsmokers showed increased sputum levels of eosinophils compared to healthy smokers. High IL-18 levels in the sputum of nonsmoking asthmatics were associated with FEV1 decline [54]. Current and ex-smokers with asthma have higher frequencies of sputum type 3 innate lymphoid cells (ILC3), which has been identified as a biomarker of airway eosinophilic inflammation, and peripheral blood CD45RO-expressing memory-like ILC3s compared with nonsmokers counterparts. ILC3 was associated with M1 alveolar macrophage and circulating neutrophil counts [55]. High levels of peripheral blood ILC2, FeNO, blood eosinophils, and serum IgE were associated with sputum eosinophil counts in eosinophilic asthmatic nonsmokers compared to healthy controls [56]. Higher serum high-sensitivity C-reactive protein (hs-CRP) levels were reported in nonsmokers with mild-to-moderate asthma than healthy controls and were associated with sputum neutrophils/eosinophils and impaired FEV1 [57]. Nonsmoking asthmatics showed higher levels of hs-CRP and blood eosinophils compared to nonsmoking healthy controls [58]. A number of inflammation biomarkers, including hs-CRP, serum total IgE, and blood/sputum eosinophils and neutrophils have been identified in higher proportions in smokers and nonsmokers with severe asthma compared to healthy nonsmokers [59]. Higher levels of Matrix metallopeptidase (MMP-12), C-X-C motif chemokine ligand-8 (CXCL8), neutrophil elastase, azurocidin 1 (AZU-1), and pro-platelet basic protein (PPBP) were observed in the sputum of ex-smoking asthmatics, which are linked to neutrophilic inflammation. Nonsmokers with asthma have significantly elevated sputum and blood eosinophil counts [60].

4.2.2. Cross-Sectional Studies

FeNO levels were reported to be high in ex-smoking and nonsmoking asthmatics. Low FeNO levels were associated with increased nitric oxide synthase (NOS2) mRNA levels in current smokers, but not in ex-smokers/nonsmokers. Current and ex-smoking asthmatics exhibit higher NADPH oxidase 2 (NOX2) mRNA levels [61]. Current smokers with severe asthma have lower FeNO value, sputum eosinophils/neutrophils, and serum-specific IgE levels than nonsmokers. Ex-smokers compared with nonsmokers have higher sputum neutrophils, blood eosinophils, and lower serum-specific IgE levels [62]. FeNO levels have been observed higher in nonsmokers than current and ex-smokers with asthma. Current and ex-smokers compared with nonsmokers had higher number of blood eosinophils [63]. FeNO values were higher in smokers and nonsmokers with uncontrolled asthma treated and/or treated with ICS than those with partly/well-controlled asthma [64]. Current smoking was associated with small airway obstruction in asthma. Interestingly, increased levels of serum IgE were associated with reduced risk of small airway obstruction in nonsmoking asthmatics compared to current and ex-smoking counterparts [65].

4.2.3. Cohort Studies

Higher FeNO levels have been observed in nonsmokers than smokers with asthma, which are associated with FEV1 and FEV1/FVC decline over 20-year follow-up [66]. In asthmatic patients with persistent obstruction where nonsmokers represented the vast majority, high sputum periostin levels were associated with FEV1 decline and high sputum eosinophil counts, resulting in increased FeNO value, blood eosinophil counts and transforming growth factor beta 1 (TGF-β1) over 2-year follow-up [67].
It can be suggested that current smoking, ex-smoking, and nonsmoking asthmatics exhibit higher levels of inflammation biomarkers, which have the potential to increase the risk. Table 3 shows the OS and inflammatory biomarkers in nonsmoking asthmatics.

5. Potential Effects of Antioxidant on CS-Induced Asthma Biomarkers

5.1. Antioxidant Vitamins

5.1.1. Vitamin A

Vitamin A (VA) derived from dietary animal-source foods has an active metabolite retinoic acid (RA), which binds retinoic acid receptors (RARs) and retinoid X receptors (RXRs) with high affinity, resulting in a regulation of ASM cell proliferation in asthma [68]. Low RA levels in ASM cells increases the severity of asthma [69]. Human ASM cells treated with RARγ-specific agonist and all-trans RA (ATRA) lead to the inhibition of platelet-derived growth factor (PDGF)-induced activator protein-1 (AP-1) regulated genes, including MMP8 and MMP9 [69]. TGF-β increases the expression of ATRA and 9-cis RA in the ASM cells of patients with severe asthma compared with those with mild-to-moderate asthma, which results in upregulation of the mRNA of β1-integrin, MMP-9, and hepatocyte growth factor receptor (HGF-R). Treatment with anti-TGF-β1 monoclonal antibody in the presence of ATRA/9-cis RA reduces the levels of MMP-9 mRNA in ASM cells. This concludes that TGF-β increases ASM cell inflammation in response to exaggerated RA receptor expression, which may lead to airway epithelial repair defects in severe asthma [70]. Administration of ATRA suppresses PDGF-induced ASM cell migration via RAR-RXR heterodimer activation and Serine-threonine kinase/Phosphatidylinositol-3 kinase (Akt/PI3K) signaling pathway inhibition [71]. Treatment with RA improves barrier strength of HBECs by reducing TNF-α and IL-6-induced airway barrier leaks and occluded din/claudin-4 [72]. ATRA treatment inhibits airway inflammation by suppressing Th2 and Th17-related cytokines (IL-4, IL-5, IL-17A), neutrophils, eosinophils, macrophages, and lymphocytes counts [73]. Administration of ATRA and 9-cis RA suppresses IL-4-induced eotaxin mRNA expression in HBECs [74]. 9-cis-RA treatment results in reversing RAR-beta (RAR-β) expression loss in the HBECs of ex-smokers, suggesting that RA may be considered as a potential agent against asthma risk in smokers [75]. Nonsmoker patients with lung emphysema treated with ATRA resulted in improvement in lung function and reduction of airway inflammation through the inhibition of TNF-α and IL-3 plasma levels [76]. Overall findings suggest that VA exerts anti-inflammatory effects on ASM/HBECs cells.

5.1.2. Carotenoids

β-carotene, also termed provitamin A/non-polar carotenoid, and other non-provitamin A/polar carotenoids (e.g., lycopene, lutein, zeaxanthin) are natural pigments, present primarily in fruits and vegetables, which have been shown to exert anti-inflammatory/OS agents for several diseases [77], including asthma [78]. It has been shown that supplementation of HBECs “BEAS-2B” with β-carotene does not promote membrane LP/lactate dehydrogenase (LDH) leakage and α-tocopherol/GSH depletion caused by gas phase smoke [79]. β-carotene exerts a protective effect in HBECs treated with CS-induced lung carcinogen benzo[a]pyrene (BaP) through increasing RAR-β expression [80].
Lycopene exerts a therapeutic effect against asthma by reducing eosinophilic infiltrates and Th2-mediated cytokines IL-4 and IL-5 production in the airways [81]. Treatment with apo-10′-lycopenoic acid, an active metabolite of lycopene, increases accumulation of nuclear factor-E2 related factor 2 (Nrf2)-mediated heme-oxygenase-1 (HO-1) activation and intracellular GSH levels and decreases intracellular ROS levels and hydrogen peroxide (H2O2)-induced LDH production in BEAS-2B [82]. Treatment of BEAS-2B with β-cryptoxanthin (BCX) reduces inflammation, as indicated by increased sirtuin1 (SIRT1) protein levels and inhibited lipopolysaccharide (LPS)-induced TNF-α, MMP2/9, IL-6, and monocyte chemoattractant protein-1 (MCP-1) mRNA levels [83]. BCX supplementation of ferrets led to inhibited CS-induced NF-kB, AP-1, and TNFα expression in the lungs [84]. These findings suggest that lycopene and BCX may protect HBECs against CS-induced inflammation and/or OS biomarkers.

5.1.3. Vitamin C and E

Epidemiological studies regarding the role of antioxidant VC and VE in the treatment of asthma have demonstrated inconsistent findings [85]. The ascorbic acid-supplemented diet has been shown to reduce the bronchoconstrictive responses in asthmatic patients, as demonstrated by decreasing post-exercise FeNO, FEV1, and urinary 9α, 11β-PGF2 levels [86]. Administration of VC to ovalbumin (OVA)-sensitized and challenged asthmatic mice attenuates airway inflammation by reducing eosinophilic infiltration into BALF [87].
VE and curcumin treatment reduces BaP-induced ROS levels by downregulating poly[ADP-ribose] polymerase 1 (PARP-1) and protein 53 (p53) activity in BEAS-2B [88]. Treatment with natural-source d-α-tocopheryl acetate increases plasma levels of α-tocopherol isoform of VE in atopic asthmatics, resulting in reduced BAL levels of IL-3 and IL-4 [89]. In allergic asthmatic adults, γ-tocopherol treatment led to suppression of LPS-induced TNFα, IL-6, and IL-1β production from peripheral blood monocytes [90]. In human and mice models, supplementation with γ-tocopherol reduced LPS-induced sputum percentages of neutrophils and eosinophils [91]. γ-tocopherol supplementation reduces sputum mucins, neutrophils, and eosinophils in mild asthmatics [92]. VE-supplemented diet results in decreased IL-4 and IL-5 levels in the murine lungs [93]. Treatment with VE attenuates AHR through reducing OS and inflammatory biomarkers. VE decreases LPS-induced IL-5, IL-13 levels, H2O2-mediated ROS production, eosinophils, neutrophils, and restores the serum activity of GSH and SOD [94]. Administration of VE reduces asthma by decreasing IL-4 levels, ROS production, serum IgE levels, and increasing GSH levels [95]. VE treatment attenuates allergic asthma by reducing levels of peroxynitrite, NO2, IgE, eotaxin, IgE, TGF-β1, IL-4, IL-5, and IL-13 [96]. Administration of supplemental α-and γ-tocopherol resulted in reduced BALF IL-5, IL-12, and IL-13 levels [97]. This suggests that VC may reduce airway inflammation, while VE may have protective effects against both OS and inflammatory biomarkers.

5.1.4. Vitamin D

Evidence from in vitro and in vivo studies has supported the protective role of VD against asthma, by which VD supplementation reduces airway inflammation and improves lung function in asthmatic patients [98]. VD treatment has been shown to decrease IL-6 and CXCL8 levels in cultured HBECs from asthmatic donors [99]. In asthmatic patients, supplementation with VD increases serum anti-inflammatory IL-10, and decreases serum levels of IgE, eosinophil, IL-5, IL-9, and IL-13 [100]. Supplementation with VD reduces asthmatic airway inflammation, as evidenced by decreased levels of IL-4 in BALF and NO2 in serum and BALF [101]. VD treatment decreases the index of airway collagen deposition, mucus reserve, and increases autophagy-related protein expression levels of hypoxia-inducible factor 1 alpha (HIF-1α) and neurogenic locus notch homolog protein 1 (Notch1), resulting in reduced airway inflammation associated with IL-6 and IL-17 cytokines [102]. Supplementation of VD reduces AHR and IgE levels in BALF and serum in asthmatic mice [103]. Administration of VD to VD-deficient mice with asthma reduces BALF levels of neutrophil, eosinophil, IL-5, and IL-13 [104]. It has been demonstrated that 1,25(OH)(2)D(3) supplementation reduces serum OVA-specific IgE levels, accompanied with increased serum levels of IL-10 via inhibition of NF-kB signaling pathway [105]. VD supplementation reduces BALF eosinophil numbers, BALF IL-6, IL-17, TNF-α levels, and increase BALF IL-10 levels [106]. VD was found to reduce serum levels of IL-6, IL-1β, TNF-α, and increase serum levels of IL-10 through downregulating high mobility group box 1 proteins (HMGB1)/TLR4/NF-κB signaling pathway [107]. Overall findings suggest that VD may have anti-inflammatory/anti-oxidants effects on HBECs due to its ability to reduce inflammatory biomarkers, which may therefore be involved in CS-induced asthma treatment.

5.2. Antioxidant Minerals

5.2.1. Iron

Iron (Fe) is a critical mineral implicated in free radical production, which has a detrimental effect on asthma, as evident by increasing plasma Fe levels in HBECs, which result in a significant increase in OS and LP [108,109]. IL-6 was shown to enhance ferroptosis in HBECs, identified as regulated cell death, by disrupting iron homeostasis and increasing ROS and MDA-dependent LP [110]. In mice sensitized to OVA, high expression of HO-1, an enzyme responsible for degrading heme into free iron, was found to be associated with airway inflammation via increased levels of IL-5, IL-13, and eosinophilia in the lung tissue [111].
High serum levels of saturation of transferrin and ferritin, as indices of Fe homeostasis, were associated with airway obstruction in smokers and nonsmokers with the lowest FEV1/FVC ratio [112]. Exposure to tobacco smoke condensate alters iron homeostasis in human respiratory epithelial cells by increasing serum Fe and ferritin accumulation in the lungs of smokers [113]. This suggests that Fe is associated with increased asthma risk and should not be recommended for smoking asthmatics.

5.2.2. Zinc, Selenium, and Copper

Evidence from in vivo and in vitro studies suggests that zinc (Zn), Se, and copper (Cu) play a significant role in reducing asthma and protecting airway epithelial cells against OS and inflammatory biomarkers [114]. Treatment of airway epithelial (HEp-2) cells with toxic copper oxide nanoparticles (CuONPs) results in induced OS by increasing ROS and 8-isoprostane production [115]. CuONPs increase AHR and the production of ROS and pro-inflammatory cytokines via activating of MAPK signaling in OVA-induced asthmatic mice [116]. Cu and Zn are key components of SOD, which results in a reduction of OS. The plasma levels of Se, Cu, Zn, and a cytosolic antioxidant enzyme, copper–zinc-superoxide dismutase (CuZnSOD), were reported to be lower in asthmatics than in healthy controls [117,118,119]. Se was found to be associated with decreased levels of OS biomarker plasma thiobarbituric acid-reactive substances (TBARS), hs-CRP levels and CD4/CD8 lymphocyte ratios [118]. An experimental study has demonstrated that Zn chelator N,N,N′,N′-tetrakis(2-pyridylmethyl)ethylenediamine inhibits TNFα-induced eotaxin mRNA expression in BEAS-2B cells [120]. Zn supplementation reduced BALF eosinophils and attenuated airway inflammation-induced solute carrier family 39 members 1 and 14 (ZIP1, ZIP14) [121]. Administration of Zn to mice with OVA-induced allergic asthma led to reduced monocytes, neutrophils, and eosinophils in BALF, MCP-1, and eotaxin protein production [122]. This suggests that Zn may have potential antioxidant effects against inflammation biomarkers.
The potential effect of antioxidant vitamins and minerals on CS-induced asthma biomarkers was shown in Table 4.

6. Concluding Remarks

CS is associated with biomarkers of OS and systemic inflammation in HBECs. Literature has shown that OS and inflammation have a significant role in the pathogenesis of asthma in current smokers, ex-smokers, and nonsmokers. There were significant differences in asthma biomarkers between smokers and nonsmokers. OS biomarkers MDA, FRAP, protein carbonyls, and peroxynitrite, and inflammation biomarkers ILC2/3, MMP-12, CXCL8, neutrophil elastase, AZU-1, and PPBP were found to associate with asthma in smokers, but not in nonsmokers. However, OS biomarkers NO2, lipid peroxide, protein sulfhydrils, and inflammatory biomarkers TNFα, IL-4, IL-37, IL-1β, IL-1RL1, IL-1R1, NLRP3, and TGF-β1 showed positive association with asthma in nonsmokers, but not in smokers. Current smokers with asthma have higher levels of OS biomarkers than nonsmokers and are thus at a heightened state of OS. The activity of the enzymatic antioxidant defense in smoking asthmatics may not adequately protect the HBECs against oxidative damage.
Evidence from a few studies suggests that dietary VC and carotenoid intake are associated with reduced asthma risk in smokers and/or nonsmokers. Supplementing VE and Se had no effects on improving lung function in smoking asthmatics.
Several in vivo and in vitro studies have demonstrated the protective effects of antioxidant vitamin and mineral against asthma biomarkers. VA, VC, BCX, and Zn might protect HBECs against inflammatory biomarkers, while VE, VD, and lycopene might provide protection against both OS and inflammatory biomarkers. Fe has adverse effects on HBECs and should be avoided for smoking and nonsmoking asthmatics.
The potential effects of antioxidant on CS-induced asthma biomarkers in smoking and nonsmoking asthmatics are difficult to determine, given a limited number of human studies. VA and carotenoids may trigger a protective effect against asthma in smokers and/or nonsmokers. However, VC, VE, VD, and Zn may have protective potential against asthma biomarkers. Such effects lead to the conclusion that these antioxidants might have beneficial effects in reducing asthma in smokers and nonsmokers, given that smoking and nonsmoking asthmatics are susceptible to CS-induced OS and inflammatory biomarkers.
The mechanisms by which antioxidant vitamins and minerals might be effective in protecting HBECs against asthma biomarkers in smokers and nonsmokers have not been fully elucidated. Human studies on the exact mechanisms (signaling pathways) linking the antioxidant intake to asthma in smokers and nonsmokers have not yet been confirmed. Few signaling pathways might be involved, as demonstrated by in vivo models (e.g., TLR4/NF-κB signaling pathway in VD). Further human studies are needed to explore the mechanisms by which antioxidants might be effective in protecting HBECs against asthma biomarkers in smokers and nonsmokers.
More studies on smokers and nonsmokers are needed to determine the associations between antioxidant intake from both diet and supplements and asthma biomarkers. Studies included in this review did not determine whether nonsmokers with asthma are affected by SHS exposure. Thus, further studies are required to examine whether antioxidant intake in nonsmoking asthmatics could protect against CS-induced asthma biomarkers.

Funding

This review received no financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

AHRAirway hyperresponsiveness
AktSerine-threonine kinase
AP-1Activator protein-1
ASMAirway smooth muscle
ATRAAll-trans RA
AZU-1Azurocidin 1
BALFBronchoalveolar Lavavge Fluid
BaPBenzo[a]pyrene
BCXβ-cryptoxanthin
COPDChronic obstructive pulmonary disease
CSCigarette smoke
CSECigarette smoke extract
CuCopper
CuONPsCopper oxide nanoparticles
CuZnSODZinc-superoxide dismutase
CXCLC-X-C motif chemokine ligand
EBCExhaled breathe condensate
ECPEosinophilic cationic protein
ERKExtracellular signal-regulated kinases
ETSEnvironmental tobacco smoke
FeIron
FeNOFractional exhaled nitric oxide
FEV1Forced expiratory volume in 1 s
FRAPFerric reducing ability of plasma
FVCForced vital capacity
GPxGlutathione peroxidase
GSHReduced glutathione
H2O2Hydrogen peroxide
HBECsHuman bronchial epithelial cells
HDACHistone deacetylase
HGFRHepatocyte growth factor receptor
HIF-1αHypoxia-inducible factor 1 alpha
HMGB1Mobility group box 1 protein
HO-1Heme-oxygenase-1
hs-CRPHigh-sensitivity C-reactive protein
ICSInhalation corticosteroid
IgEImmunoglobulin E
ILInterleukin
ILCInnate lymphoid cell
IL-18R1Interleukin 18 receptor 1
IL-1RL1Interleukin 1 receptor-like 1
8-iso-PGFIsoprostane-8-iso prostaglandin F
JNKc-Jun N-terminal kinase
LABALong-acting β2 adrenergic
LCLung cancer
LDHLactate dehydrogenase
LPLipid peroxidation
LPSLipopolysaccharide
MAPKsMitogen-activated protein kinases
MCP-1Monocyte chemoattractant protein-1
MDAMalondialdehyde
MMPMatrix metallopeptidases
MPOMyeloperoxidase
α7nAChRα7 nicotinic acetylcholine receptor
NF-κBNuclear transcription factor-kappaB
NLRPNLR Family CARD Domain Containing
NNKNitrosamine 4(methylnitrosamino)-1-(3–pyridyl)-1-butanone
NO2Nitrite
NOSNitric oxide synthase
Notch1Neurogenic locus notch homolog protein 1
NOX2NADPH oxidase 2
Nrf2Nuclear factor-E2 related factor 2
OSOxidative stress
OVAOvalbumin
OXSR1Oxidative stress responsive kinase 1
P53Protein 53
PARP-1Poly[ADP-ribose] polymerase 1
PDGFPlatelet-derived growth factor
PEFRPeak expiratory flow rate
PGF2Prostaglandin F2
PI3KPhosphatidylinositol-3 kinase
PPBPPro-platelet basic protein
QoLQuality of life
RARetinoic acid
RARsRetinoic acid receptors
RDBPCRandomized double blind placebo control
RXRsRetinoid X receptors
RCTsRandomised controlled trials
ROSReactive oxygen species
SeSelenium
SHSSecondhand smoke
SIRT1Sirtuin1
SLC-39Solute carrier family 39
SODSuperoxide dismutase
TBARSThiobarbituric acid reactive substances
TGF-β1Transforming growth factor beta 1
ThT-helper
TLRToll-like receptors
TNF-αTumor necrosis factor α
TNFRSF11ATNF receptor superfamily member 11a
VAVitamin A
VCVitamin C
VDVitamin D
VEVitamin E
ZnZinc

References

  1. Gan, H.; Hou, X.; Zhu, Z.; Xue, M.; Zhang, T.; Huang, Z.; Cheng, Z.J.; Sun, B. Smoking: A leading factor for the death of chronic respiratory diseases derived from Global Burden of Disease Study 2019. BMC Pulm. Med. 2022, 22, 149. [Google Scholar] [CrossRef] [PubMed]
  2. Institute for Health Metrics and Evaluation. Global Burden of Disease 2017. 2017. Available online: http://vizhub.healthdata.org/gbd-compare/# (accessed on 6 March 2023).
  3. Aoshiba, K.; Nagai, A. Differences in airway remodeling between asthma and chronic obstructive pulmonary disease. Clin. Rev. Allergy Immunol. 2004, 27, 35–43. [Google Scholar] [CrossRef]
  4. Doeing, D.C.; Solway, J. Airway smooth muscle in the pathophysiology and treatment of asthma. J. Appl. Physiol. 2013, 114, 834–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kume, H. Role of airway smooth muscle in inflammation related to asthma and COPD. Adv. Exp. Med. Biol. 2021, 1303, 139–172. [Google Scholar] [PubMed]
  6. Shimoda, T.; Obase, Y.; Kishikawa, R.; Iwanaga, T. Influence of cigarette smoking on airway inflammation and inhaled corticosteroid treatment in patients with asthma. Allergy Asthma Proc. 2016, 37, 50–58. [Google Scholar] [CrossRef] [PubMed]
  7. Tommola, M.; Ilmarinen, P.; Tuomisto, L.E.; Haanpää, J.; Kankaanranta, T.; Niemelä, O.; Kankaanranta, H. The effect of smoking on lung function: A clinical study of adult-onset asthma. Eur. Respir. J. 2016, 48, 1298–1306. [Google Scholar] [CrossRef] [Green Version]
  8. Korsbæk, N.; Landt, E.M.; Dahl, M. Second-hand smoke exposure associated with risk of respiratory symptoms, asthma, and COPD in 20,421 adults from the General Population. J. Asthma Allergy 2021, 14, 1277–1284. [Google Scholar] [CrossRef]
  9. Keogan, S.; Alonso, T.; Sunday, S.; Tigova, O.; Fernández, E.; López, M.J.; Gallus, S.; Semple, S.; Tzortzi, A.; Boffi, R.; et al. Lung function changes in patients with chronic obstructive pulmonary disease (COPD) and asthma exposed to secondhand smoke in outdoor areas. J. Asthma 2021, 58, 1169–1175. [Google Scholar] [CrossRef]
  10. Coogan, P.F.; Castro-Webb, N.; Yu, J.; O’Connor, G.T.; Palmer, J.R.; Rosenberg, L. Active and passive smoking and the incidence of asthma in the Black Women’s Health Study. Am. J. Respir. Crit. Care Med. 2015, 191, 168–176. [Google Scholar] [CrossRef] [Green Version]
  11. Van der Toorn, M.; Rezayat, D.; Kauffman, H.F.; Bakker, S.J.L.; Gans, R.O.B.; Koëter, G.H.; Choi, A.M.K.; Van Oosterhout, A.J.M.; Slebos, D.-J. Lipid-soluble components in cigarette smoke induce mitochondrial production of reactive oxygen species in lung epithelial cells. Am. J. Physiol. Lung. Cell Mol. Physiol. 2009, 297, L109–L114. [Google Scholar] [CrossRef] [Green Version]
  12. Zhou, G.; Xiao, W.; Xu, C.; Hu, Y.; Wu, X.; Huang, F.; Lu, X.; Shi, C.; Wu, X. Chemical constituents of tobacco smoke induce the production of interleukin-8 in human bronchial epithelium, 16HBE cells. Tob. Induc. Dis. 2016, 14, 24. [Google Scholar] [CrossRef] [Green Version]
  13. Ong, J.; van den Berg, A.; Faiz, A.; Boudewijn, I.M.; Timens, W.; Vermeulen, C.J.; Oliver, B.G.; Kok, K.; Terpstra, M.M.; van den Berge, M.; et al. Current smoking is associated with decreased expression of miR-335–5p in parenchymal lung fibroblasts. Int. J. Mol. Sci. 2019, 20, 5176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mirra, D.; Cione, E.; Spaziano, G.; Esposito, R.; Sorgenti, M.; Granato, E.; Cerqua, I.; Muraca, L.; Iovino, P.; Gallelli, L.; et al. Circulating microRNAs expression profile in lung inflammation: A preliminary study. J. Clin. Med. 2022, 11, 5446. [Google Scholar] [CrossRef] [PubMed]
  15. Cipollina, C.; Bruno, A.; Fasola, S.; Cristaldi, M.; Patella, B.; Inguanta, R.; Vilasi, A.; Aiello, G.; La Grutta, S.; Torino, C.; et al. Cellular and molecular signatures of oxidative stress in bronchial epithelial cell models injured by cigarette smoke extract. Int. J. Mol. Sci. 2022, 23, 1770. [Google Scholar] [CrossRef] [PubMed]
  16. 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] [PubMed] [Green Version]
  17. Alsharairi, N.A. Scutellaria baicalensis and their natural flavone compounds as potential medicinal drugs for the treatment of nicotine-induced non-small-cell lung cancer and asthma. Int. J. Environ. Res. Public Health 2021, 18, 5243. [Google Scholar] [CrossRef] [PubMed]
  18. Vonk, J.M.; Scholtens, S.; Postma, D.S.; Moffatt, M.F.; Jarvis, D.; Ramasamy, A.; Wjst, M.; Omenaas, E.R.; Bouzigon, E.; Demenais, F.; et al. Adult onset asthma and interaction between genes and active tobacco smoking: The GABRIEL consortium. PLoS ONE 2017, 12, e0172716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Uh, S.-T.; Park, J.-S.; Koo, S.-M.; Kim, Y.-K.; Kim, K.U.; Kim, M.-A.; Shin, S.-W.; Son, J.-H.; Park, H.-W.; Shin, H.D.; et al. Association of genetic variants of NLRP4 with exacerbation of asthma: The effect of smoking. DNA Cell. Biol 2019, 38, 76–84. [Google Scholar] [CrossRef]
  20. Losol, P.; Kim, S.H.; Ahn, S.; Lee, S.; Choi, J.-P.; Kim, Y.-H.; Hong, S.-J.; Kim, B.-S.; Chang, Y.-S. Genetic variants in the TLR-related pathway and smoking exposure alter the upper airway microbiota in adult asthmatic patients. Allergy 2021, 76, 3217–3220. [Google Scholar] [CrossRef]
  21. Kim, M.-H.; Chang, H.S.; Lee, J.-U.; Shim, J.-S.; Park, J.-S.; Cho, Y.-J.; Park, C.-S. Association of genetic variants of oxidative stress responsive kinase 1 (OXSR1) with asthma exacerbations in non-smoking asthmatics. BMC Pulm. Med. 2022, 22, 3. [Google Scholar] [CrossRef]
  22. Belcaro, G.; Luzzi, R.; Cesinaro Di Rocco, P.; Cesarone, M.R.; Dugall, M.; Feragalli, B.; Errichi, B.M.; Ippolito, E.; Grossi, M.G.; Hosoi, M.; et al. Pycnogenol® improvements in asthma management. Panminerva. Med. 2011, 53, 57–64. [Google Scholar] [PubMed]
  23. Thomson, N.C.; Spears, M. The influence of smoking on the treatment response in patients with asthma. Curr. Opin. Allergy Clin. Immunol. 2005, 5, 57–63. [Google Scholar] [CrossRef] [PubMed]
  24. Thomson, N.C.; Shepherd, M.; Spears, M.; Chaudhuri, R. Corticosteroid insensitivity in smokers with asthma: Clinicalevidence, mechanisms, and management. Treat Respir. Med. 2006, 5, 467–481. [Google Scholar] [CrossRef] [PubMed]
  25. Chatkin, J.M.; Dullius, C.R. The management of asthmatic smokers. Asthma Res. Pract. 2016, 2, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Duman, B.; Borekci, S.; Akdeniz, N.; Gazioglu, S.B.; Deniz, G.; Gemicioglu, B. Inhaled corticosteroids’ effects on biomarkers in exhaled breath condensate and blood in patients newly diagnosed with asthma who smoke. J. Asthma 2022, 59, 1613–1620. [Google Scholar] [CrossRef] [PubMed]
  27. Alsharairi, N.A. The effects of dietary supplements on asthma and lung cancer risk in smokers and non-smokers: A review of the literature. Nutrients 2019, 11, 725. [Google Scholar] [CrossRef] [Green Version]
  28. Alsharairi, N.A. Supplements for smoking-related lung diseases. Encyclopedia 2021, 1, 76–86. [Google Scholar] [CrossRef]
  29. Alsharairi, N.A. Dietary antioxidants and lung cancer risk in smokers and non-smokers. Healthcare 2022, 10, 2501. [Google Scholar] [CrossRef]
  30. Santillan, A.A.; Camargo, C.A., Jr.; Colditz, G.A. A meta-analysis of asthma and risk of lung cancer (United States). Cancer Causes Control. 2003, 14, 327–334. [Google Scholar] [CrossRef]
  31. Rosenberger, A.; Bickeböller, H.; McCormack, V.; Brenner, D.R.; Duell, E.J.; Tjønneland, A.; Friis, S.; Muscat, J.E.; Yang, P.; Wichmann, H.E.; et al. Asthma and lung cancer risk: A systematic investigation by the International Lung Cancer Consortium. Carcinogenesis 2012, 33, 587–597. [Google Scholar] [CrossRef] [Green Version]
  32. Burns, J.S.; Dockery, D.W.; Neas, L.M.; Schwartz, J.; Coull, B.A.; Raizenne, M.; Speizer, F.E. Low dietary nutrient intakes and respiratory health in adolescents. Chest 2007, 132, 238–245. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, W.; Li, W.; Du, J. Association between dietary carotenoid intakes and the risk of asthma in adults: A cross-sectional study of NHANES, 2007–2012. BMJ Open 2022, 12, e052320. [Google Scholar] [CrossRef] [PubMed]
  34. Pearson, P.J.K.; Lewis, S.A.; Britton, J.; Fogarty, A. Vitamin E supplements in asthma: A parallel group randomised placebo controlled trial. Thorax 2004, 59, 652–656. [Google Scholar] [CrossRef] [Green Version]
  35. Shaheen, S.O.; Newson, R.B.; Rayman, M.P.; Wong, A.P.-L.; Tumilty, M.K.; Phillips, J.M.; Potts, J.F.; Kelly, F.J.; White, P.T.; Burney, P.G.J. Randomised, double blind, placebo-controlled trial of selenium supplementation in adult asthma. Thorax 2007, 62, 483–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Van der Vaart, H.; Postma, D.S.; Timens, W.; Ten Hacken, N.H.T. Acute effects of cigarette smoke on inflammation and oxidative stress: A review. Thorax 2004, 59, 713–721. [Google Scholar] [CrossRef] [Green Version]
  37. Yadav, A.S.; Saini, M. Evaluation of systemic antioxidant level and oxidative stress in relation to lifestyle and disease progression in asthmatic patients. J. Med. Biochem. 2016, 35, 55–62. [Google Scholar] [CrossRef] [Green Version]
  38. Bartoli, M.L.; Novelli, F.; Costa, F.; Malagrinò, L.; Melosini, L.; Bacci, E.; Cianchetti, S.; Dente, F.L.; Di Franco, A.; Vagaggini, B.; et al. Malondialdehyde in exhaled breath condensate as a marker of oxidative stress in different pulmonary diseases. Mediators Inflamm. 2011, 2011, 891752. [Google Scholar] [CrossRef] [PubMed]
  39. Anes, A.B.; Nasr, H.B.; Fetoui, H.; Bchir, S.; Chahdoura, H.; Yacoub, S.; Garrouch, A.; Benzarti, M.; Tabka, Z.; Chahed, K. Alteration in systemic markers of oxidative and antioxidative status in Tunisian patients with asthma: Relationships with clinical severity and airflow limitation. J. Asthma 2016, 53, 227–237. [Google Scholar] [CrossRef]
  40. Mak, J.C.W.; Leung, H.C.M.; Ho, S.P.; Law, B.K.W.; Lam, W.K.; Tsang, K.W.T.; Ip, M.C.M.; Chan-Yeung, M. Systemic oxidative and antioxidative status in Chinese patients with asthma. J. Allergy Clin. Immunol. 2004, 114, 260–264. [Google Scholar] [CrossRef]
  41. Nadeem, A.; Chhabra, S.K.; Masood, A.; Raj, H.G. Increased oxidative stress and altered levels of antioxidants in asthma. J. Allergy Clin. Immunol. 2003, 111, 72–78. [Google Scholar] [CrossRef]
  42. Deveci, F.; Ilhan, N.; Turgut, T.; Akpolat, N.; Kirkil, G.; Muz, M.H. Glutathione and nitrite in induced sputum from patients with stable and acute asthma compared with controls. Ann. Allergy Asthma Immunol. 2004, 93, 91–97. [Google Scholar] [CrossRef] [PubMed]
  43. Hillas, G.; Kostikas, K.; Mantzouranis, K.; Bessa, V.; Kontogianni, K.; Papadaki, G.; Papiris, S.; Alchanatis, M.; Loukides, S.; Bakakos, P. Exhaled nitric oxide and exhaled breath condensate pH as predictors of sputum cell counts in optimally treated asthmatic smokers. Respirology 2011, 16, 811–818. [Google Scholar] [CrossRef] [PubMed]
  44. Michils, A.; Louis, R.; Peché, R.; Baldassarre, S.; Van Muylem, A. Exhaled nitric oxide as a marker of asthma control in smoking patients. Eur. Respir. J. 2009, 33, 1295–1301. [Google Scholar] [CrossRef]
  45. Pavlidis, S.; Takahashi, K.; Kwong, F.N.K.; Xie, J.; Hoda, U.; Sun, K.; Elyasigomari, V.; Agapow, P.; Loza, M.; Baribaud, F.; et al. “T2-high” in severe asthma related to blood eosinophil, exhaled nitric oxide and serum periostin. Eur. Respir. J. 2019, 53, 1800938. [Google Scholar] [CrossRef]
  46. Chamitava, L.; Cazzoletti, L.; Ferrari, M.; Garcia-Larsen, V.; Jalil, A.; Degan, P.; Fois, A.G.; Zinellu, E.; Fois, S.S.; Pasini, A.M.F.; et al. Biomarkers of oxidative stress and inflammation in chronic airway diseases. Int. J. Mol. Sci. 2020, 21, 4339. [Google Scholar] [CrossRef] [PubMed]
  47. Malinovschi, A.; Backer, V.; Harving, H.; Porsbjerg, C. The value of exhaled nitric oxide to identify asthma in smoking patients with asthma-like symptoms. Respir. Med. 2012, 106, 794–801. [Google Scholar] [CrossRef] [Green Version]
  48. Zietkowski, Z.; Tomasiak-Lozowska, M.M.; Skiepko, R.; Zietkowska, E.; Bodzenta-Lukaszyk, A. Eotaxin-1 in exhaled breath condensate of stable and unstable asthma patients. Respir. Res. 2010, 11, 110. [Google Scholar] [CrossRef] [Green Version]
  49. Krisiukeniene, A.; Babusyte, A.; Stravinskaite, K.; Lotvall, J.; Sakalauskas, R.; Sitkauskiene, B. Smoking affects eotaxin levels in asthma patients. J. Asthma 2009, 46, 470–476. [Google Scholar] [CrossRef]
  50. Rossios, C.; Pavlidis, S.; Hoda, U.; Kuo, C.-H.; Wiegman, C.; Russell, K.; Sun, K.; Loza, M.J.; Baribaud, F.; Durham, A.L.; et al. Sputum transcriptomics reveal upregulation of IL-1 receptor family members in patients with severe asthma. J. Allergy Clin. Immunol. 2018, 141, 560–570. [Google Scholar] [CrossRef] [Green Version]
  51. Chalmers, G.W.; MacLeod, K.J.; Thomson, L.; Little, S.A.; McSharry, C.; Thomson, N.C. Smoking and airway inflammation in patients with mild asthma. Chest 2001, 120, 1917–1922. [Google Scholar] [CrossRef]
  52. Thomson, N.C.; Chaudhuri, R.; Spears, M.; Haughney, J.; McSharry, C. Serum periostin in smokers and never smokers with asthma. Respir. Med. 2015, 109, 708–7015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yildiz, H.; Alp, H.H.; Sünnetçioğlu, A.; Ekin, S.; Çilingir, B.M. Evaluation serum levels of YKL-40, Periostin, and some inflammatory cytokines together with IL-37, a new anti-inflammatory cytokine, in patients with stable and exacerbated asthma. Heart Lung 2021, 50, 177–183. [Google Scholar] [CrossRef]
  54. Rovina, N.; Dima, E.; Gerassimou, C.; Kollintza, A.; Gratziou, C.; Roussos, C. IL-18 in induced sputum and airway hyperresponsiveness in mild asthmatics: Effect of smoking. Respir. Med. 2009, 103, 1919–1925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ham, J.; Kim, J.; Sohn, K.-H.; Park, I.-W.; Choi, B.-W.; Chung, D.H.; Cho, S.-H.; Kang, H.R.; Jung, J.-W.; Kim, H.Y. Cigarette smoke aggravates asthma by inducing memory-like type 3 innate lymphoid cells. Nat. Commun. 2022, 13, 3852. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, T.; Wu, J.; Zhao, J.; Wang, J.; Zhang, Y.; Liu, L.; Cao, L.; Liu, Y.; Dong, L. Type 2 innate lymphoid cells: A novel biomarker of eosinophilic airway inflammation in patients with mild to moderate asthma. Respir. Med. 2015, 109, 1391–1396. [Google Scholar] [CrossRef] [Green Version]
  57. Shimoda, T.; Obase, Y.; Kishikawa, R.; Iwanaga, T. Serum high-sensitivity C-reactive protein can be an airway inflammation predictor in bronchial asthma. Allergy Asthma Proc. 2015, 36, e23–e28. [Google Scholar] [CrossRef]
  58. Halvani, A.; Tahghighi, F.; Nadooshan, H.H. Evaluation of correlation between airway and serum inflammatory markers in asthmatic patients. Lung. India 2012, 29, 143–146. [Google Scholar] [CrossRef]
  59. Mikus, M.S.; Kolmert, J.; Andersson, L.I.; Östling, J.; Knowles, R.G.; Gómez, C.; Ericsson, M.; Thörngren, J.-O.; Khoonsari, P.E.; Dahlén, B.; et al. Plasma proteins elevated in severe asthma despite oral steroid use and unrelated to Type-2 inflammation. Eur. Respir. J. 2022, 59, 2100142. [Google Scholar] [CrossRef]
  60. Takahashi, K.; Pavlidis, S.; Kwong, F.N.K.; Hoda, U.; Rossios, C.; Sun, K.; Loza, M.; Baribaud, F.; Chanez, P.; Fowler, S.J.; et al. Sputum proteomics and airway cell transcripts of current and ex-smokers with severe asthma in U-BIOPRED: An exploratory analysis. Eur. Respir. J. 2018, 51, 1702173. [Google Scholar] [CrossRef]
  61. Emma, R.; Bansal, A.T.; Kolmert, J.; Wheelock, C.E.; Dahlen, S.-E.; Loza, M.J.; De Meulder, B.; Lefaudeux, D.; Auffray, C.; Dahlen, B.; et al. Enhanced oxidative stress in smoking and ex-smoking severe asthma in the U-BIOPRED cohort. PLoS ONE 2018, 13, e0203874. [Google Scholar] [CrossRef] [Green Version]
  62. Thomson, N.C.; Chaudhuri, R.; Heaney, L.G.; Bucknall, C.; Niven, R.M.; Brightling, C.E.; Menzies-Gow, A.N.; Mansur, A.H.; McSharry, C. Clinical outcomes and inflammatory biomarkers in current smokers and exsmokers with severe asthma. J. Allergy Clin. Immunol. 2013, 131, 1008–1016. [Google Scholar] [CrossRef] [PubMed]
  63. Giovannelli, J.; Chérot-Kornobis, N.; Hulo, S.; Ciuchete, A.; Clément, G.; Amouyel, P.; Matran, R.; Dauchet, L. Both exhaled nitric oxide and blood eosinophil count were associated with mild allergic asthma only in non-smokers. Clin. Exp. Allergy 2016, 46, 543–554. [Google Scholar] [CrossRef] [PubMed]
  64. Kostikas, K.; Papaioannou, A.I.; Tanou, K.; Giouleka, P.; Koutsokera, A.; Minas, M.; Papiris, S.; Gourgoulianis, K.I.; Taylor, D.R.; Loukides, S. Exhaled NO and exhaled breath condensate pH in the evaluation of asthma control. Respir. Med. 2011, 105, e526–e532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chu, S.; Ma, L.; Wei, J.; Wang, J.; Xu, Q.; Chen, M.; Jiang, M.; Luo, M.; Wu, J.; Mai, L.; et al. Smoking status modifies the relationship between Th2 biomarkers and small airway obstruction in asthma. Can. Respir. J. 2021, 2021, 1918518. [Google Scholar] [CrossRef]
  66. Nerpin, E.; Ferreira, D.S.; Weyler, J.; Schlunnsen, V.; Jogi, R.; Semjen, C.R.; Gislasson, T.; Demoly, P.; Heinrich, J.; Nowak, D.; et al. Bronchodilator response and lung function decline: Associations with exhaled nitric oxide with regard to sex and smoking status. World Allergy Organ. J. 2021, 14, 100544. [Google Scholar] [CrossRef]
  67. Cianchetti, S.; Cardini, C.; Puxeddu, I.; Latorre, M.; Bartoli, M.L.; Bradicich, M.; Dente, F.; Bacci, E.; Celi, A.; Paggiaro, P. Distinct profile of inflammatory and remodelling biomarkers in sputum of severe asthmatic patients with or without persistent airway obstruction. World Allergy Organ. J. 2019, 12, 100078. [Google Scholar] [CrossRef]
  68. Timoneda, J.; Rodríguez-Fernández, L.; Zaragozá, R.; Marín, M.P.; Cabezuelo, M.T.; Torres, L.; Viña, J.R.; Barber, T. Vitamin A deficiency and the lung. Nutrients 2018, 10, 1132. [Google Scholar] [CrossRef] [Green Version]
  69. Defnet, A.E.; Shah, S.D.; Huang, W.; Shapiro, P.; Deshpande, D.A.; Kane, M.A. Dysregulated retinoic acid signaling in airway smooth muscle cells in asthma. FASEB J. 2021, 35, e22016. [Google Scholar] [CrossRef]
  70. Druilhe, A.; Zahm, J.-M.; Benayoun, L.; El Mehdi, D.; Grandsaigne, M.; Dombret, M.-C.; Mosnier, I.; Feger, B.; Depondt, J.; Aubier, M.; et al. Epithelium expression and function of retinoid receptors in asthma. Am. J. Respir. Cell. Mol. Biol. 2008, 38, 276–282. [Google Scholar] [CrossRef]
  71. Day, R.M.; Lee, Y.H.; Park, A.-M.; Suzuki, Y.J. Retinoic acid inhibits airway smooth muscle cell migration. Am. J. Respir. Cell. Mol. Biol. 2006, 34, 695–703. [Google Scholar] [CrossRef]
  72. Callaghan, P.J.; Rybakovsky, E.; Ferrick, B.; Thomas, S.; Mullin, J.M. Retinoic acid improves baseline barrier function and attenuates TNF-α-induced barrier leak in human bronchial epithelial cell culture model, 16HBE 14o. PLoS ONE 2020, 15, e0242536. [Google Scholar] [CrossRef] [PubMed]
  73. Wu, J.; Zhang, Y.; Liu, Q.; Zhong, W.; Xia, Z. All-trans retinoic acid attenuates airway inflammation by inhibiting Th2 and Th17 response in experimental allergic asthma. BMC Immunol. 2013, 14, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Takamura, K.; Nasuhara, Y.; Kobayashi, M.; Betsuyaku, T.; Tanino, Y.; Kinoshita, I.; Yamaguchi, E.; Matsukura, S.; Schleimer, R.P.; Nishimura, M. Retinoic acid inhibits interleukin-4-induced eotaxin production in a human bronchial epithelial cell line. Am. J. Physiol. Lung. Cell. Mol. Physiol. 2004, 286, L777–L785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kurie, J.M.; Lotan, R.; Lee, J.J.; Lee, J.S.; Morice, R.C.; Liu, D.D.; Xu, X.-C.; Khuri, F.R.; Ro, J.Y.; Hittelman, W.N.; et al. Treatment of former smokers with 9-cis-retinoic acid reverses loss of retinoic acid receptorbeta expression in the bronchial epithelium: Results from a randomized placebo-controlled trial. J. Natl. Cancer. Inst. 2003, 95, 206–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Frankenberger, M.; Heimbeck, I.; Möller, W.; Mamidi, S.; Kaßner, G.; Pukelsheim, K.; Wjst, M.; Neiswirth, M.; Kroneberg, P.; Lomas, D.; et al. Inhaled all-trans retinoic acid in an individual with severe emphysema. Eur. Respir. J. 2009, 34, 1487–1489. [Google Scholar] [CrossRef] [Green Version]
  77. Saini, R.K.; Prasad, P.; Lokesh, V.; Shang, X.; Shin, J.; Keum, Y.-S.; Lee, J.-H. Carotenoids: Dietary sources, extraction, encapsulation, bioavailability, and health benefits-A review of recent advancements. Antioxidants 2022, 11, 795. [Google Scholar] [CrossRef]
  78. Park, H.S.; Kim, S.R.; Kim, J.O.; Lee, Y.C. The roles of phytochemicals in bronchial asthma. Molecules 2010, 15, 6810–6834. [Google Scholar] [CrossRef]
  79. Arora, A.; Willhite, C.A.; Liebler, D.C. Interactions of beta-carotene and cigarette smoke in human bronchial epithelial cells. Carcinogenesis 2001, 22, 1173–1178. [Google Scholar] [CrossRef] [Green Version]
  80. Prakash, P.; Liu, C.; Hu, K.-Q.; Krinsky, N.I.; Russell, R.M.; Wang, X.-D. Beta-carotene and beta-apo-14′-carotenoic acid prevent the reduction of retinoic acid receptor beta in benzo[a]pyrene-treated normal human bronchial epithelial cells. J. Nutr. 2004, 134, 667–673. [Google Scholar] [CrossRef] [Green Version]
  81. Hazlewood, L.C.; Wood, L.G.; Hansbro, P.M.; Foster, P.S. Dietary lycopene supplementation suppresses Th2 responses and lung eosinophilia in a mouse model of allergic asthma. J. Nutr. Biochem. 2011, 22, 95–100. [Google Scholar] [CrossRef]
  82. Lian, F.; Wang, X.-D. Enzymatic metabolites of lycopene induce Nrf2-mediated expression of phase II detoxifying/antioxidant enzymes in human bronchial epithelial cells. Int. J. Cancer 2008, 123, 1262–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chiaverelli, R.A.; Hu, K.-Q.; Liu, C.; Lim, J.Y.; Daniels, M.S.; Xia, H.; Mein, J.; von Lintig, J.; Wang, X.-D. β-Cryptoxanthin attenuates cigarette-smoke-induced lung lesions in the absence of carotenoid cleavage enzymes (BCO1/BCO2) in mice. Molecules 2023, 28, 1383. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, C.; Bronson, R.T.; Russell, R.M.; Wang, X.-D. β-Cryptoxanthin supplementation prevents cigarette smoke-induced lung inflammation, oxidative damage, and squamous metaplasia in ferrets. Cancer Prev. Res. 2011, 4, 1255–1266. [Google Scholar] [CrossRef] [Green Version]
  85. Allen, S.; Britton, J.R.; Leonardi-Bee, J.A. Association between antioxidant vitamins and asthma outcome measures: Systematic review and meta-analysis. Thorax 2009, 64, 610–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tecklenburg, S.L.; Mickleborough, T.D.; Fly, A.D.; Bai, Y.; Stager, J.M. Ascorbic acid supplementation attenuates exercise-induced bronchoconstriction in patients with asthma. Respir. Med. 2007, 101, 1770–1778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Chang, H.-H.; Chen, C.-S.; Lin, J.-Y. High dose vitamin C supplementation increases the Th1/Th2 cytokine secretion ratio, but decreases eosinophilic infiltration in bronchoalveolar lavage fluid of ovalbumin-sensitized and challenged mice. J. Agric. Food Chem. 2009, 57, 10471–10476. [Google Scholar] [CrossRef]
  88. Zhu, W.; Cromie, M.M.; Cai, Q.; Lv, T.; Singh, K.; Gao, W. Curcumin and vitamin E protect against adverse effects of benzo[a]pyrene in lung epithelial cells. PLoS ONE 2014, 9, e92992. [Google Scholar] [CrossRef] [Green Version]
  89. Hoskins, A.; Roberts, J.L.; Milne, G.; Choi, L.; Dworski, R. Natural-source d-α-tocopheryl acetate inhibits oxidant stress and modulates atopic asthma in humans In Vivo. Allergy 2012, 67, 676–682. [Google Scholar] [CrossRef] [Green Version]
  90. Wiser, J.; Alexis, N.E.; Jiang, Q.; Wu, W.; Robinette, C.; Roubey, R.; Peden, D.B. In vivo gamma-tocopherol supplementation decreases systemic oxidative stress and cytokine responses of human monocytes in normal and asthmatic subjects. Free Radic. Biol. Med. 2008, 45, 40–49. [Google Scholar] [CrossRef] [Green Version]
  91. Hernandez, M.L.; Wagner, J.G.; Kala, A.; Mills, K.; Wells, H.B.; Alexis, N.E.; Lay, J.C.; Jiang, Q.; Zhang, H.; Zhou, H.; et al. Vitamin E, γ-tocopherol, reduces airway neutrophil recruitment after inhaled endotoxin challenge in rats and in healthy volunteers. Free Radic. Biol. Med. 2013, 60, 56–62. [Google Scholar] [CrossRef] [Green Version]
  92. Burbank, A.J.; Duran, C.G.; Pan, Y.; Burns, P.; Jones, S.; Jiang, Q.; Yang, C.; Jenkins, S.; Wells, H.; Alexis, N.; et al. Gamma tocopherol-enriched supplement reduces sputum eosinophilia and endotoxin-induced sputum neutrophilia in volunteers with asthma. J. Allergy Clin. Immunol. 2018, 141, 1231–1238.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Talati, M.; Meyrick, B.; Peebles Jr, R.S.; Davies, S.S.; Dworski, R.; Mernaugh, R.; Mitchell, D.; Boothby, M.; Roberts, L.J.; Sheller, J.R. Oxidant stress modulates murine allergic airway responses. Free Radic. Biol. Med. 2006, 40, 1210–1219. [Google Scholar] [CrossRef] [PubMed]
  94. Quoc, Q.L.; Bich, T.C.T.; Kim, S.-H.; Park, H.-S.; Shin, Y.S. Administration of vitamin E attenuates airway inflammation through restoration of Nrf2 in a mouse model of asthma. J. Cell Mol. Med. 2021, 25, 6721–6732. [Google Scholar] [CrossRef]
  95. Li, J.; Li, L.; Chen, H.; Chang, Q.; Liu, X.; Wu, Y.; Wei, C.; Li, R.; Kwan, J.K.C.; Yeung, K.L.; et al. Application of vitamin E to antagonize SWCNTs-induced exacerbation of allergic asthma. Sci. Rep. 2014, 4, 4275. [Google Scholar] [CrossRef] [Green Version]
  96. Mabalirajan, U.; Aich, J.; Leishangthem, G.D.; Sharma, S.K.; Dinda, A.K.; Ghosh, B. Effects of vitamin E on mitochondrial dysfunction and asthma features in an experimental allergic murine model. J. Appl. Physiol. 2009, 107, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
  97. McCary, C.A.; Abdala-Valencia, H.; Berdnikovs, S.; Cook-Mills, J.M. Supplemental and highly elevated tocopherol doses differentially regulate allergic inflammation: Reversibility of α-tocopherol and γ-tocopherol’s effects. J. Immunol. 2011, 186, 3674–3685. [Google Scholar] [CrossRef] [Green Version]
  98. Hall, S.C.; Agrawal, D.K. Vitamin D and bronchial asthma: An overview of data from the past 5 years. Clin. Ther. 2017, 39, 917–929. [Google Scholar] [CrossRef] [Green Version]
  99. Pfeffer, P.E.; Lu, H.; Mann, E.H.; Chen, Y.-H.; Ho, T.-R.; Cousins, D.J.; Corrigan, C.; Kelly, F.J.; Mudway, I.S.; Hawrylowicz, C.M. Effects of vitamin D on inflammatory and oxidative stress responses of human bronchial epithelial cells exposed to particulate matter. PLoS ONE 2018, 13, e0200040. [Google Scholar] [CrossRef] [Green Version]
  100. Ramos-Martínez, E.; López-Vancell, M.R.; Fernández de Córdova-Aguirre, J.C.; Rojas-Serrano, J.; Chavarría, A.; Velasco-Medina, A.; Velázquez-Sámano, G. Reduction of respiratory infections in asthma patients supplemented with vitamin D is related to increased serum IL-10 and IFNγ levels and cathelicidin expression. Cytokine 2018, 108, 239–246. [Google Scholar] [CrossRef]
  101. Adam-Bonci, T.-I.; Bonci, E.-A.; Pârvu, A.-E.; Herdean, A.-I.; Moț, A.; Taulescu, M.; Ungur, A.; Pop, R.-M.; Bocșan, C.; Irimie, A. Vitamin D supplementation: Oxidative stress modulation in a mouse model of ovalbumin-induced acute asthmatic airway inflammation. Int. J. Mol. Sci. 2021, 22, 7089. [Google Scholar] [CrossRef]
  102. Huang, C.; Peng, M.; Tong, J.; Zhong, X.; Xian, J.; Zhong, L.; Deng, J.; Huang, Y. Vitamin D ameliorates asthma-induced lung injury by regulating HIF-1α/Notch1 signaling during autophagy. Food Sci. Nutr. 2022, 10, 2773–2785. [Google Scholar] [CrossRef] [PubMed]
  103. Fischer, K.D.; Hall, S.C.; Agrawal, D.K. Vitamin D supplementation reduces induction of epithelial-mesenchymal transition in allergen sensitized and challenged mice. PLoS ONE 2016, 11, e0149180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Gorman, S.; Weeden, C.E.; Tan, D.H.W.; Scott, N.M.; Hart, J.; Foong, R.E.; Mok, D.; Stephens, N.; Zosky, G.; Hart, P.H. Reversible control by vitamin D of granulocytes and bacteria in the lungs of mice: An ovalbumin-induced model of allergic airway disease. PLoS ONE 2013, 8, e67823. [Google Scholar] [CrossRef]
  105. Taher, Y.A.; van Esch, B.C.A.M.; Hofman, G.A.; Henricks, P.A.J.; van Oosterhout, A.J.M. 1alpha,25-dihydroxyvitamin D3 potentiates the beneficial effects of allergen immunotherapy in a mouse model of allergic asthma: Role for IL-10 and TGF-beta. J. Immunol. 2008, 180, 5211–5221. [Google Scholar] [CrossRef] [Green Version]
  106. Agrawal, T.; Gupta, G.K.; Agrawal, D.K. Vitamin D supplementation reduces airway hyperresponsiveness and allergic airway inflammation in a murine model. Clin. Exp. Allergy 2013, 43, 672–683. [Google Scholar] [PubMed]
  107. Zhang, H.; Yang, N.; Wang, T.; Dai, B.; Shang, Y. Vitamin D reduces inflammatory response in asthmatic mice through HMGB1/TLR4/NF-κB signaling pathway. Mol. Med. Rep. 2018, 17, 2915–2920. [Google Scholar]
  108. Ekmekci, O.P.; Donma, O.; Sardoğan, E.; Yildirim, N.; Uysal, O.; Demirel, H.; Demir, T. Iron, nitric oxide, and myeloperoxidase in asthmatic patients. Biochemistry 2004, 69, 462–467. [Google Scholar] [CrossRef]
  109. Narula, M.K.; Ahuja, G.K.; Whig, J.; Narang, A.P.S.; Soni, R.K. Status of lipid peroxidation and plasma iron level in bronchial asthmatic patients. Indian J. Physiol. Pharmacol. 2007, 51, 289–292. [Google Scholar]
  110. Han, F.; Li, S.; Yang, Y.; Bai, Z. Interleukin-6 promotes ferroptosis in bronchial epithelial cells by inducing reactive oxygen species-dependent lipid peroxidation and disrupting iron homeostasis. Bioengineered 2021, 12, 5279–5288. [Google Scholar] [CrossRef]
  111. Kuribayashi, K.; Iida, S.-I.; Nakajima, Y.; Funaguchi, N.; Tabata, C.; Fukuoka, K.; Fujimori, Y.; Ihaku, D.; Nakano, T. Suppression of heme oxygenase-1 activity reduces airway hyperresponsiveness and inflammation in a mouse model of asthma. J. Asthma 2015, 52, 662–668. [Google Scholar] [CrossRef]
  112. Ghio, A.J.; Hilborn, E.D. Indices of iron homeostasis correlate with airway obstruction in an NHANES III cohort. Int. J. Chron. Obstruct. Pulmon. Dis. 2017, 12, 2075–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ghio, A.J.; Hilborn, E.D.; Stonehuerner, J.G.; Dailey, L.A.; Carter, J.D.; Richards, J.H.; Crissman, K.M.; Foronjy, R.F.; Uyeminami, D.L.; Pinkerton, K.E. Particulate matter in cigarette smoke alters iron homeostasis to produce a biological effect. Am. J. Respir. Crit. Care. Med. 2008, 178, 1130–1138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zajac, D. Mineral micronutrients in asthma. Nutrients 2021, 13, 4001. [Google Scholar] [CrossRef] [PubMed]
  115. Fahmy, B.; Cormier, S.A. Copper oxide nanoparticles induce oxidative stress and cytotoxicity in airway epithelial cells. Toxicol. Vitr. 2009, 23, 1365–1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Park, J.-W.; Lee, I.-C.; Shin, N.-R.; Jeon, C.-M.; Kwon, O.-K.; Ko, J.-W.; Kim, J.-C.; Oh, S.-R.; Shin, I.-S.; Ahn, K.-S. Copper oxide nanoparticles aggravate airway inflammation and mucus production in asthmatic mice via MAPK signaling. Nanotoxicology 2016, 10, 445–452. [Google Scholar] [CrossRef] [PubMed]
  117. Sagdic, A.; Sener, O.; Bulucu, F.; Karadurmus, N.; Özel, H.E.; Yamanel, L.; Tasci, C.; Naharci, I.; Ocal, R. Aydin Oxidative stress status and plasma trace elements in patients with asthma or allergic rhinitis. Allergol. Immunopathol. 2011, 39, 200–205. [Google Scholar] [CrossRef] [PubMed]
  118. Guo, C.-H.; Liu, P.-J.; Hsia, S.; Chuang, C.-J.; Chen, P.-C. Role of certain trace minerals in oxidative stress, inflammation, CD4/CD8 lymphocyte ratios and lung function in asthmatic patients. Ann. Clin. Biochem. 2011, 48, 344–351. [Google Scholar] [CrossRef]
  119. Mao, S.; Wu, L.; Shi, W. Association between trace elements levels and asthma susceptibility. Respir. Med. 2018, 145, 110–119. [Google Scholar] [CrossRef]
  120. Richter, M.; Cantin, A.M.; Beaulieu, C.; Cloutier, A.; Larivée, P. Zinc chelators inhibit eotaxin, RANTES, and MCP-1 production in stimulated human airway epithelium and fibroblasts. Am. J. Physiol. Lung Cell Mol. Physiol. 2003, 285, L719–L729. [Google Scholar] [CrossRef]
  121. Lang, C.; Murgia, C.; Leong, M.; Tan, L.-W.; Perozzi, G.; Knight, D.; Ruffin, R.; Zalewski, P. Anti-inflammatory effects of zinc and alterations in zinc transporter mRNA in mouse models of allergic inflammation. Am. J. Physiol. Lung. Cell. Mol. Physiol. 2007, 292, L577–L584. [Google Scholar] [CrossRef] [Green Version]
  122. Lu, H.; Xin, Y.; Tang, Y.; Shao, G. Zinc suppressed the airway inflammation in asthmatic rats: Effects of zinc on generation of eotaxin, MCP-1, IL-8, IL-4, and IFN-γ. Biol. Trace. Elem. Res. 2012, 150, 314–321. [Google Scholar] [CrossRef] [PubMed]
Table 1. Antioxidants and asthma risk in relation to smoking status.
Table 1. Antioxidants and asthma risk in relation to smoking status.
DesignStudy PopulationAntioxidantsMain FindingsRef.
Cross-sectionalTotal subjects = 2112 12th grade US studentsVC, VE (diet)Low dietary VC intake (<110 mg/day) was associated with FEV1 decline and respiratory symptoms in smokers with asthma[32]
Smokers = 515VE intake was not associated with asthma
Cross-sectionalTotal subjects = 13,039 US adults (20–80 yrs)Total carotenoids (diet and supplement)High intake of carotenoids (≥165.59 μg/kg/day) was associated with reduced asthma risk in nonsmokers (OR = 0.63, 95% CI = 0.42 to 0.93), current smokers (OR = 0.54, 95% CI = 0.36 to 0.83), and ex-smokers (OR = 0.64, 95% CI = 0.42 to 0.97)[33]
Current asthma = 1784; non-current asthma = 11,255
Nonsmokers= 7106; current smokers= 3304; ex-smokers= 2624
RDBPCTotal subjects = 72 UK nonsmoking asthmatics (18–60 yrs)VE (supplement)VE had no beneficial effects on asthma[34]
500 mg VE capsules (D-α-tocopherol) in soya bean oil or matched placebo (capsules, gelatine base) for 6 weeks
RDBPCTotal subjects = 197 UK smoking and nonsmoking asthmatics (18–54 yrs)Se (supplement)Plasma Se was increased by 48% in the Se group. However, no significant improvement in QoL score was observed in the Se group compared with placebo[35]
100 μg/day high-Se yeast preparation or matched placebo (yeast only) for 24 weeks
Abbreviations: RDBPC, randomized double blind placebo control; VC, vitamin C; VE, vitamin E; Se, selenium; QoL, quality of life; OR, odds ratio.
Table 2. OS biomarkers in smoking and nonsmoking asthmatics.
Table 2. OS biomarkers in smoking and nonsmoking asthmatics.
DesignStudy PopulationOS BiomarkersRef.
Case-control studyTotal subjects = 210 Indian (13–80 yrs) Smokers/nonsmokers (asthmatics = 19/101; healthy controls = 29/61)Asthmatic smokers = MDA ↑, FRAP ↓[37]
Case-control studyTotal subjects = 194 Italian patients with different pulmonary diseases (average 45.8 yrs)Asthmatic current smokers = MDA ↑[38]
Asthmatics (current and ex-smokers) = 64; healthy controls (nonsmokers) = 14
Case-control studyTotal subjects = 329 Tunisian adults (average 43.6 yrs)Asthmatic current smokers = Protein carbonyls, peroxynitrite ↑[39]
Asthmatic current smokers/healthy controls = 14/73; Asthmatic ex- smokers/healthy controls = 17/13
Asthmatic nonsmokers/healthy controls = 120/92
Case-control studyTotal subjects = 266 Chinese adults (39–47 yrs)Asthmatic smokers and nonsmokers = SOD, GSH ↑[40]
Asthmatic smokers/nonsmokers = 25/106; healthy controls (nonsmokers) = 135
Case-control studyTotal subjects = 61 Indian (15–40 yrs) Asthmatic nonsmokers/healthy controls= 38/23Asthmatic nonsmokers = SOD, NO2, protein carbonyls, lipid peroxide ↑[41]
GPx, protein sulfhydrils ↓
Case-control studyTotal subjects = 32 Turkish adults (average 41 yrs)Asthmatic nonsmokers = GSH, NO2[42]
Stable asthmatic nonsmokers = 11; Severe asthmatic nonsmokers = 10; Healthy nonsmokers = 11
(↓) decrease, (↑) increase.
Table 3. Inflammatory biomarkers in smoking and nonsmoking asthmatics.
Table 3. Inflammatory biomarkers in smoking and nonsmoking asthmatics.
DesignStudy PopulationInflammation BiomarkersRef.
Case-control studyTotal subjects = 143 Greek adults (average 48.7 yrs)Asthmatic smokers = FeNo ↓, eosinophils ↑[43]
Asthmatic smokers/nonsmokers = 40/43
Healthy smokers/nonsmokers = 30/30
Case-control studyTotal subjects = 470 Belgium adults (average 41 yrs)Asthmatic smokers = FeNo ↓[44]
Asthmatic smokers/nonsmokers = 59/411
Case-control studyTotal subjects = 147 European adults (average 46.8 yrs)Asthmatic smokers = FeNo ↓[45]
Asthmatic smokers = 18; Severe Asthmatic nonsmokers = 49; Mild-moderate asthmatic nonsmokers = 36; healthy nonsmoker = 44Asthmatic nonsmokers = FeNo, eosinophil ↑
Case-control studyTotal subjects = 1230 Italian adults (20–65 yrs)Asthmatic nonsmokers = FeNo, eosinophils, neutrophils ↑[46]
Current/past asthmatic nonsmokers = 404/185; Current and past chronic bronchitis smokers = 92; healthy controls = 549
Case-control studyTotal subjects = 282 Danish (14–44 yrs)Asthmatic nonsmokers = FeNo, ↑[47]
Asthmatic current/ex-smokers = 112/62; Asthmatic nonsmokers = 108
Case-control studyTotal subjects = 58 Polish adults (25–45 yrs)Asthmatic nonsmokers = FeNo, eotaxin, eosinophil ↑[48]
Asthmatic nonsmokers/Healthy controls = 46/12
Case-control studyTotal subjects = 68 Lithuanian adults (average 55.2 yrs)Asthmatic smokers = Eotaxin, neutrophils, eosinophils ↑[49]
Asthmatic smokers/nonsmokers = 19/26; Healthy smokers and non-smokers = 23Asthmatic nonsmokers = Eotaxin, neutrophils, eosinophils, IL-5 ↑
Case-control studyTotal subjects = 86 UK adults (average 50 yrs)Asthmatic smokers = IL-1β, IL-5, IL-18R1 ↑[50]
Severe asthmatic smokers/nonsmokers = 21/37; mild-moderate asthmatics = 15; healthy controls = 13Asthmatic nonsmokers = Eotaxin, IL-4, IL-5, IL-1β, IL-1RL1, IL-1R1, NLRP3 ↑
Case-control studyTotal subjects = 97 UK adults (average 37 yrs)Asthmatic smokers = eosinophils, ECP, neutrophils, IL-8 ↑[51]
Asthmatic smokers/nonsmokers = 31/36; Nonasthmatic smokers/nonsmokers = 15/15Asthmatic nonsmokers = eosinophils, ECP ↑
Case-control studyTotal subjects = 152 UK adults (18–75 yrs)Asthmatic smokers = Periostin ↓[52]
Asthmatic smokers/nonsmokers = 56/51; Healthy smokers/nonsmokers = 20/25Asthmatic nonsmokers = Periostin ↑
Case-control studyTotal subjects = 89 Turkish adults (25–65 yrs)Asthmatic nonsmokers = Periostin, TNFα, IL-4, IL-5, YKL-40 ↑, IL-37 ↓[53]
Asthmatic nonsmokers/healthy controls = 59/30
Case-control studyTotal subjects = 79 Greek adults (average 46 yrs)Asthmatic smokers = eosinophils, neutrophils ↑, IL-18 ↓[54]
Asthmatic smokers/nonsmokers = 24/22; Healthy smokers/nonsmokers = 16/17Asthmatic nonsmokers = eosinophils, neutrophils, IL-18 ↑
Case-control studyTotal subjects = 115 Korean adults (average 55 yrs)Asthmatic smokers = ILC3, eosinophils, neutrophils ↑[55]
Asthmatic smokers/nonsmokers = 58/33; Healthy smokers/nonsmokers = 11/13
Case-control studyTotal subjects = 168 Chinese adults (average 36 yrs)Eosinophilic asthmatic nonsmokers = ILC2, IgE, eosinophils, FeNO ↑[56]
Eosinophilic asthmatic/non asthmatic nonsmokers = 62/64; Healthy controls = 42
Case-control studyTotal subjects = 85 Japanese adults (20–60 yrs)Asthmatic nonsmokers = hs-CRP, eosinophils, neutrophils ↑[57]
Asthmatic nonsmokers/healthy controls = 45/40
Case-control studyTotal subjects = 98 Iranian adults (average 35 yrs)Asthmatic nonsmokers = hs-CRP, eosinophils ↑[58]
Asthmatic nonsmokers receiving/not receiving inhaled fluticasone (500 µg/day) = 31/30; Healthy controls = 37
Case-control studyTotal subjects = 525 European adults (36–55 yrs)Asthmatic smokers and nonsmokers = hs-CRP, eosinophils, neutrophils, IgE ↑[59]
Smokers or ex-smokers with severe asthma = 95; Nonsmokers with severe asthma = 263; Nonsmokers with mild-moderate asthma = 76; healthy nonsmoker = 91
Case-control studyTotal subjects = 88 European adults (39–50 yrs)Asthmatic ex-smokers = MMP-12, CXCL8, neutrophil elastase, AZU-1, PPBP ↑[60]
Asthmatic current smokers = 11; Asthmatic ex-smokers = 22; Asthmatic nonsmoker s = 37; Healthy nonsmoker = 18Asthmatic nonsmokers = eosinophil ↑
Cross-sectional studyTotal subjects = 324 European adults with severe asthma (average 52.5 yrs)Asthmatic current smokers = FeNo ↓, NOS2, NOx2[61]
Current smokers = 42; Ex-smokers = 112; Nonsmokers = 260Asthmatic ex-smokers = FeNo, NOX2
Asthmatic nonsmokers = FeNo ↑
Cross-sectional studyTotal subjects = 740 UK patients with severe asthma (6–43 yrs)Asthmatic current smokers = FeNO, blood eosinophils, sputum eosinophils[62]
Current smokers = 69; Ex-smokers = 210; Nonsmokers = 461sputum neutrophils, IgE ↓
Asthmatic ex-smokers = FeNO, sputum neutrophils, blood eosinophils ↑, IgE ↓
Asthmatic nonsmokers = FeNO, IgE, sputum neutrophils, sputum eosinophils, blood eosinophils ↑
Cross-sectional studyTotal subjects = 1578 French patients with asthma (40–64 yrs)Asthmatic current and ex- smokers = FeNO ↓, blood eosinophils ↑[63]
Current smokers = 294; Ex-smokers = 473; Nonsmokers = 812Asthmatic nonsmokers = FeNO ↑, blood eosinophils ↓
Cross-sectional studyTotal subjects = 274 Greek patients with asthma (average 50 yrs)Asthmatic smokers and nonsmokers = FeNO ↑[64]
Inhaled corticosteroid (ICS)-treated smokers = 50; ICS-untreated smokers = 32; ICS-treated nonsmokers = 144; ICS-untreated nonsmokers = 48
Cross-sectional studyTotal subjects = 478 Chinese patients with asthma (average 45 yrs)Asthmatic nonsmokers = IgE ↑[65]
Obstructive group (current smokers = 70; ex-smokers = 44; nonsmokers = 271), Normal group (current smokers = 9; ex-smokers = 6; nonsmokers = 78)
Prospective cohort studyTotal subjects = 4257 European and Australian adults (average 54 yrs)Asthmatic nonsmokers = FeNO ↑[66]
Current asthma (smokers = 97; nonsmokers = 554)
Non-asthma (smokers = 651; nonsmokers = 2955)
Prospective cohort studyTotal subjects = 45 Italian adults with severe asthma (average 60 yrs)Asthmatic nonsmokers = FeNO, periostin, eosinophil, TGF-β1 ↑[67]
Nonsmokers = 42; ex-smokers = 3
(↓) decrease, (↑) increase.
Table 4. Potential influence of antioxidant vitamins and minerals on CS-induced asthma biomarkers.
Table 4. Potential influence of antioxidant vitamins and minerals on CS-induced asthma biomarkers.
Study DesignAntioxidant Vitamins/MineralsConcentration/Supplement IntakeOS BiomarkersInflammation BiomarkersRef.
In vitroVA (RARγ, ATRA)1–1000 μM-MMPs (MMP8, MMP9) ↓[69]
In vitroVA (ATRA, 9-cis RA)1 mM in the presence of 0.2 mg/mL neutralizing anti-TGF-β1-MMP9 ↓[70]
In vitroVA (RA)5, 15, 50 μM-TNFα, IL-6 ↓[72]
In vivoVA (ATRA)400 μg/mL-IL-4, IL-5, IL-17A, neutrophils, eosinophils ↓[73]
In vitroVA (ATRA, 9-cis RA)10−6–10−10 M-IL-4, eotaxin ↓[74]
In vivoLycopene0.16 mg (equivalent to 8 mg/kg per day) plus 0.05 mg VE and 0.006 mg β-carotene-IL-4, IL-5, eosinophils ↓[81]
In vitroLycopene3, 5 10 μMGSH ↑, ROS, H2O2-[82]
In vivo/vivoBCX1–4 μM (in vitro)
30−43 nmol BCX/g liver (in vivo)
-Neutrophils, TNFα, IL-6, MMPs (MMP2, MMP9) ↓[83]
RCTVC1500 mg/day for 2 weeks, or a placebo-FeNO ↓[86]
In vivoVC130 mg VC/kg bw/day-Eosinophils ↓[87]
Non-randomized trial in vivoVE1500 IU/day for 16 weeks-IL-3, IL-4 ↓[89]
ex-vivoVE1200 mg/day for 8 days-TNFα, IL-1β, IL-6 ↓[90]
RDBPCVE1200 mg/day for 14 days, or a placebo-Eosinophils, neutrophils ↓[92]
In vivoVE50 mg VE/kg bw/dayH2O2, ROS ↓, GSH, SOD ↑IL-5, IL-13, eosinophils, neutrophils ↓,[94]
In vivoVE0.2 and 2.0 mg VE/kg bw/dayROS ↓, GSH ↑IL-4, IgE ↓[95]
In vivoVE5, 10, 15, and 20 IU VE/kg bw/dayNO2, peroxynitrite ↓IL-4, IL-5, IL-13, eotaxin, IgE, TGF-β1 ↓[96]
In vivoVE0.2 or 2 mg VE/kg bw/day-IL-5, IL-12, IL-13 ↓[97]
In vitroVD100 μM-IL-6, CXCL8 ↓[99]
RCTVD0.25 μg/day calcitriol for 6 months, or a placebo-IL-5, IL-9, IL-13, IgE, eosinophils ↓, IL-10 ↑[100]
In vivoVD50 μg/kg VD/kg bw/dayNO2IL-4 ↓[101]
In vivoVD100, 500, or 1000 IU VD/kg bw/day-IL-6, IL-17 ↓[102]
In vivoVD10,000 IU VD/kg bw/day-IgE ↓[103]
In vivoVD2280 IU VD/kg bw/day-IL-5, IL-13, eosinophils, neutrophils ↓[104]
In vivoVD1 mg VD/kg bw/day-IgE ↓, IL-10 ↑[105]
In vivoVD10,000 IU VD/kg bw/day-IL-6, IL-17, TNFα, eosinophils ↓, IL-10 ↑[106]
In vivoVD1 μg/mL/20 g VD/kg bw/day-IL-6, IL-1β, TNFα, ↓, IL-10 ↑[107]
In vitroFe3.3 M plus ferroptosis inhibitor Fer-1 (0.1 μM)MDA, ROS ↑, GSH ↓IL-6 ↑[110]
In vitroZn2 μM-TNFα, eotaxin ↓[120]
In vivoZn0, 54, or 100 μg Zn/kg bw/day-Eosinophils ↓[121]
In vivoZn95 mg Zn/kg bw/day-Eosinophils, neutrophils, eotaxin ↓[122]
Abbreviations: RDBPC, randomized double blind placebo control; RCT, randomized controlled trial; VA, vitamin A; ATRA, All-trans RA; RA, retinoic acid; RAR, retinoic acid receptors; BCX, β-cryptoxanthin; VC, vitamin C; VE, vitamin E; VD, vitamin D; Fe, iron; Zn, zinc. (↓) decrease, (↑) increase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alsharairi, N.A. Antioxidant Intake and Biomarkers of Asthma in Relation to Smoking Status—A Review. Curr. Issues Mol. Biol. 2023, 45, 5099-5117. https://doi.org/10.3390/cimb45060324

AMA Style

Alsharairi NA. Antioxidant Intake and Biomarkers of Asthma in Relation to Smoking Status—A Review. Current Issues in Molecular Biology. 2023; 45(6):5099-5117. https://doi.org/10.3390/cimb45060324

Chicago/Turabian Style

Alsharairi, Naser A. 2023. "Antioxidant Intake and Biomarkers of Asthma in Relation to Smoking Status—A Review" Current Issues in Molecular Biology 45, no. 6: 5099-5117. https://doi.org/10.3390/cimb45060324

Article Metrics

Back to TopTop