Next Article in Journal
Comparison of Ferric Sodium EDTA in Combination with Vitamin C, Folic Acid, Copper Gluconate, Zinc Gluconate, and Selenomethionine as Therapeutic Option for Chronic Kidney Disease Patients with Improvement in Inflammatory Status
Next Article in Special Issue
The Effect of Palmitoylethanolamide on Pain Intensity, Central and Peripheral Sensitization, and Pain Modulation in Healthy Volunteers—A Randomized, Double-Blinded, Placebo-Controlled Crossover Trial
Previous Article in Journal
Vitamin D Status Presents Different Relationships with Severity in Metabolic-Associated Fatty Liver Disease Patients with or without Hepatitis B Infection
Previous Article in Special Issue
Gut Microbiome and Organ Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Zinc in the Pathogenesis of Lung Disease

1
Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
2
Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
3
Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA 94305, USA
4
Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
5
Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
6
Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
7
Department of Thoracic Surgery, Shengjing Hospital, China Medical University, Shenyang 110022, China
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(10), 2115; https://doi.org/10.3390/nu14102115
Submission received: 1 April 2022 / Revised: 10 May 2022 / Accepted: 16 May 2022 / Published: 19 May 2022

Abstract

:
Lung diseases, such as asthma, chronic obstructive pulmonary diseases (COPD), and cystic fibrosis (CF), are among the leading causes of mortality and morbidity globally. They contribute to substantial economic burdens on society and individuals. Currently, only a few treatments are available to slow the development and progression of these diseases. Thus, there is an urgent unmet need to develop effective therapies to improve quality of life and limit healthcare costs. An increasing body of clinical and experimental evidence suggests that altered zinc and its regulatory protein levels in the systemic circulation and in the lungs are associated with these disease’s development and progression. Zinc plays a crucial role in human enzyme activity, making it an essential trace element. As a cofactor in metalloenzymes and metalloproteins, zinc involves a wide range of biological processes, such as gene transcription, translation, phagocytosis, and immunoglobulin and cytokine production in both health and disease. Zinc has gained considerable interest in these lung diseases because of its anti-inflammatory, antioxidant, immune, and metabolic modulatory properties. Here we highlight the role and mechanisms of zinc in the pathogenesis of asthma, COPD, CF, acute respiratory distress syndrome, idiopathic pulmonary fibrosis, and pulmonary hypertension.

1. Background

Zinc is a critical nutritional trace element required for the growth and development of almost all known organisms. Zinc plays crucial roles in maintaining cellular, molecular, and systemic biological processes such as cell proliferation, differentiation, apoptosis, DNA and RNA synthesis, RBC production, tissue maintenance, immune function, glucose and lipid metabolism, and cell signaling pathways [1,2,3]. Zinc is required for the function and structure of about 2500 proteins, accounting for about 10% of all human proteins, including important enzymes, cytokines, transcription factors, and other proteins [4]. Zinc acts as a structural, catalytic, and regulatory signaling molecule. About 1.4–2.3 g of zinc is found in a healthy human adult, 85% of which resides in muscle tissues and bones, 11% in skin, and 4% in all other tissues. In plasma or serum, zinc exists as bound to albumin (60%), alpha-macroglobulin (30%), and transferrin (10%). Under normal physiological conditions, zinc level is tightly regulated in the body through a coordinated and combined carrier-mediated mechanism during absorption, transport, and secretion processes. Zinc is primarily taken up in the small intestine from foods. The efficiency of zinc absorption is influenced by several factors, including zinc status in the body and zinc concentration in the diet. The zinc “importers” ZIP 1–14, the zinc exporters ZnT 1–10, and numerous binding proteins maintain zinc homeostasis in the body [5].
Zinc has long been seen as a “two-edged sword” in the body, as both too high and too low zinc levels have been related to negative consequences in cells and tissues. There has been evidence of low zinc levels in a number of conditions, including asthma, lung cancer, diabetes, hypertension, and autoimmune and other inflammatory diseases [6,7,8,9]. Low zinc levels promote apoptosis in systemic endothelium and respiratory epithelium, while high zinc levels can also induce apoptosis in cultured cerebral and pulmonary endothelia [10]. On the other hand, low zinc levels have been demonstrated to prevent apoptosis in systemic endothelial cells triggered by cadmium, linoleic acid, and tumor necrosis factor alpha [10]. High zinc levels can also increase the production of reactive oxygen species, which induces oxidative stress in the lungs. Many epidemiological, clinical, and experimental evidence suggests that aberrant levels of zinc and zinc regulatory proteins are associated with the development and/or progression of several lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), coronavirus disease 2019 (COVID-19), acute respiratory distress syndrome (ARDS), pulmonary hypertension (PH), and respiratory infectious diseases (Figure 1, Table 1). Several state-of-the-art reviews have already summarized current knowledge of the role of zinc and zinc-regulatory molecules in COVID-19 [11,12,13,14,15] and different respiratory tract infections [16,17]. Thus, in this review, we do not focus on COVID-19 and different respiratory tract infections; instead, we highlight current knowledge of the role of zinc and zinc-regulatory proteins in asthma, COPD, IPF, CF, ARDS, and PH.

2. Regulation of Zinc Levels in the Lung

The respiratory epithelium is now widely recognized as the lung’s first line of defense against a wide range of exogenous and endogenous insults. In the respiratory epithelium, zinc plays an important role in preventing death receptor–mediated apoptosis and barrier failure. Zinc can also function as an anti-inflammatory, an antioxidant, an anti-apoptotic molecule, an organelle stabilizer, a critical wound healing component, and a cofactor for DNA synthesis in respiratory epithelium [38]. Bao and Knoell demonstrated that zinc depletion increases apoptosis and causes degradation of junction proteins, loss of cell-to-cell contact, and enhanced epithelial permeability [39]. In a study of rats fed a zinc-deficient diet, the diet was shown to decrease bronchial cilia length, number of cilia, and number of cilia per cell, while zinc supplementation affected the integrity of the bronchial epithelium. Apart from airway epithelial cells, zinc is also known to exert immunomodulatory effects and antimicrobial effects on monocytes, including alveolar macrophages [40]. Human monocytes and airway epithelium have been reported to have an important role for zinc, ZIP8, and NFkB [41]. Chronic alcohol intake has been shown to reduce zinc levels and the functionality of alveolar macrophages, particularly phagocytic activity, in humans and rats [42,43].

3. The Role of Zinc Dyshomeostasis in Lung Disease

3.1. Asthma

Asthma is an inflammatory disease of the airways and lungs that can develop at any age and affects approximately 10% of the population in developed nations [44]. The disease is characterized by aberrant responses to otherwise harmless environmental stimuli, causing chronic inflammation, smooth muscle contraction, and mucus hypersecretion in the airways. This results in airway narrowing, which leads to airflow obstruction and breathing difficulties [44]. Numerous studies have demonstrated a significant decrease in zinc levels in the serum of asthma patients [18,45,46,47,48,49,50,51,52,53]. However, some studies did not find this decrease in serum zinc levels in asthma patients [54,55,56,57]. Zinc levels were also low in the sputum [58], saliva [59], and nails [60], but not in the erythrocytes [61], of asthmatic patients. An asthma-related symptom, wheezing, was linked with low zinc content [62,63,64]. There was also a significant negative relationship between serum zinc levels and total IgE and a positive relationship between zinc levels and FEV1 [18]. In a murine model of ovalbumin-induced allergic airway inflammation in the context of low zinc condition, zinc deficiency was shown to induce oxidative damage and airway epithelial cell apoptosis [19]. Mice fed a zinc-deficient diet had worse asthma symptoms, such as increased airway hyper-responsiveness, airway inflammation, and epithelial apoptosis, compared to mice fed a normal diet [19].
An appropriate zinc level in the body is crucial for maintaining the balance between anti- and pro-oxidative and anti- and pro-inflammatory responses that are disrupted in asthma. As low zinc levels have been linked to asthma in preclinical studies, zinc supplementation or the promotion of zinc homeostasis would be a new treatment option for asthma patients. The researchers found that zinc supplementation with drinking water reduced the number of eosinophils in the bronchoalveolar lavage fluid (BALF) of mice treated with ovalbumin [65]. Zinc supplementation has been reported to improve asthma symptoms in asthma patients [20]. In addition, appropriate zinc consumption during pregnancy was linked with a reduced risk of wheezing among newborns [66]. Zinc supplementation by inhalation, however, may lead to allergic inflammation. For instance, in allergic and non-allergic mouse models, ZnoNP increased airway inflammation [67,68]. Zinc supplementation for asthma needs further evaluation and optimization.

3.2. COPD

COPD is one of the leading causes of mortality and morbidity globally, with a significant social and economic burden [69,70,71,72]. COPD is a complex disease of the airways and lungs characterized by chronic airway inflammation associated with alveolar destruction (i.e., emphysema), airway remodeling, and irreversible airflow limitation. The most common cause of COPD is cigarette smoking; however, nonsmokers can also develop the disease. Currently, COPD is incurable, but some treatments can reduce the symptoms and slow the progression of the disease.
Many clinical and experimental studies have shown that dysregulation of zinc and zinc-associated proteins is linked with COPD pathogenesis [22,24,73,74,75,76,77]. Clinically, smokers with low dietary zinc intake exhibit a significantly higher prevalence of COPD [73]. Notably, insufficient dietary zinc intake was found to be common in patients with COPD [74,75,76]. However, it is unclear how insufficient dietary zinc intake leads to pulmonary dysfunction in smokers and COPD. There have also been numerous studies demonstrating that low zinc levels associated with COPD and cigarette smoke are linked with airflow obstruction, oxidative stress, inflammation, apoptosis, DNA damage, and risk of infections, allergens, and cancer [73,76,78,79,80,81,82,83]. Moreover, randomized prospective clinical trials of dietary and oral nutritional supplements improved several clinically relevant functional outcomes, such as total intake, anthropometric measures, grip strength, inspiratory and expiratory muscle strength, and exercise performance in patients with COPD [84,85]. These findings suggest that nutritional supplements may be effective in treating COPD.
A study recently showed a high incidence of low zinc levels in forty-one rural Midwestern veterans with COPD and agricultural dust exposures [21]. This study also showed that zinc-deficient mice show more severe airway inflammation in response to repeated agriculture dust exposure for 21 days. Likewise, to determine the effect of low zinc intake on chronic cigarette smoke (CS) exposure–induced experimental COPD, Knoell et al. conducted experiments on mice fed a moderate zinc-deficient diet or transgenic Zip8 knockout and overexpressing mice exposed to either room air or cigarette smoke [22]. The authors showed that mice fed a restricted zinc diet have significantly increased CS-induced emphysema-like alveolar enlargement compared to mice fed a normal diet. Additionally, even when mice were given the normal zinc diet, Zip8 depletion or overexpression showed worse lung damage in mice exposed to chronic CS exposure. The results from this study support the idea of developing micronutrient-based therapies that can boost zinc levels and identify a vital zinc transporter, Zip8, that is important in maintaining balance within the lung microenvironment by protecting it from CS-induced lung damage. However, it is unclear from the study how increased and decreased Zip8 expression induced the prolonged CS-induced alveolar tissue loss. In the future, it would be critical to determine how zip8 and zinc dyshomeostasis affects lung cellular functions, inflammation, ROS production, and chemokine production in the context of CS exposure.
Hamon et al. measured zinc concentrations in the bronchoalveolar lavage (BAL) supernatant of 20 healthy controls, 17 healthy smokers, and 20 current and 19 ex-smoker COPD subjects [23]. The authors showed that zinc concentrations in the bronchoalveolar lavage fluid of smokers and patients with COPD are significantly decreased and positively associated with alveolar macrophage efferocytosis [23], a process by which phagocytic cells remove apoptotic cells. Further in vitro studies revealed that zinc chelator TPEN treatment significantly decreased efferocytosis in macrophages. The authors then showed a significant reduction in efferocytosis ability and intracellular zinc levels in the macrophages isolated from zinc transporter Zip1 null mice, indicating that ZIP1 could play a critical role in the macrophage efferocytosis process. They also demonstrated that zinc chelation to mimic zinc deficiency increases ZIP2 expression while no change took place in ZIP1 expression in the human THP-1 macrophage cell line, suggesting that in macrophages, zinc homeostasis is maintained by zinc transporters ZIP1 and ZIP2 differently responded to zinc deficiency. Moreover, using a CS-induced mouse model of COPD, a human ex vivo air–liquid interface model, and human lung tissues from COPD patients with and without a smoking history, the same research team also demonstrated an uncoupling role of zinc trafficking and autophagy in airway epithelial cells that play a vital role and that could be a therapeutic target in COPD [24].

3.3. CF

CF is a chronic and progressive genetic disease linked with mutations in the CFTR gene and defective chloride transport across the epithelial cell membranes. The clinical symptoms of CF include a thick, dry mucus that obstructs the airways, persistent pulmonary infections, bronchiectasis, pancreatic insufficiency, and increased chloride levels in sweat.
Several studies have shown a dysregulation of zinc levels (lower in blood and higher in sputum) in adults and children with CF [25,58,86,87,88,89]. In the first three years of life, low serum zinc contents were detected in one-third of children with CF [86]. This study also found a discrepant link between growth and serum zinc in cross-sectional and longitudinal analyses; further study to better understand the role of zinc in growth in children with CF is warranted. A low-plasma zinc concentration was reported in adults with CF and moderate lung disease who had good nutritional status, and that low zinc level was linked with worse clinical outcomes [25]. There is variable success with oral zinc supplements in CF [90,91,92]. A recent exciting study showed that zinc deficiency through the unique splicing switch of ZIP2 contributes to CF-associated MUC5AC hypersecretion in airway epithelial cells [26]. Importantly, evidence from both in vivo and in vitro experiments of the CF model suggests that mucosal iron supplementation can restore airway epithelial cells’ chloride secretion by stimulating calcium-dependent chloride channels [27]. In addition to the potential therapeutic value of this finding, if zinc supplements have to be applied to mucosal surfaces of the airways to be effective, it would be imperative to develop efficient and safe methods of delivering zinc into the airways. While zinc is a relatively non-toxic metal when administered as an oral supplement, its direct administration into the airways can cause several side effects, including olfactory loss and inflammation of the respiratory tract accompanied by bronchial hyperresponsiveness [93,94]. Additionally, several recent reports showed that airborne particulate matter contains significant quantities of zinc, leading to sensitization to common aeroallergens and the occurrence or exacerbation of respiratory or allergic conditions [95,96,97].

3.4. IPF

Although extensive research has been conducted on pulmonary fibrosis, how pulmonary fibrosis occurs remains largely unclear, which has caused the unfortunate lack of effective treatments for patients suffering from progressive pulmonary fibrosis. IPF is a chronic and most common form of interstitial lung disease. The etiology and pathogenesis of IPF are unknown. IPF is characterized by repeated epithelial cell injuries and insufficient alveolar epithelium repair, which leads to excessive fibroblast activation and lung fibrosis [98,99]. The clinical symptoms of IPF include cough, dyspnea, and increasing immobility [100]. IPF primarily affects patients over the age of 60. It has a poor prognosis, with a median survival time ranging between 3 and 5 years [100,101,102]. The incidence rate for IPF has greatly differed by geography; North America and eastern Europe have experienced the highest prevalence, averaging between 3 and 9 new cases per 100,000 person-years, while East Asia had the lowest, averaging fewer than 4 new cases per 100,000 person-years [103,104]. While the etiology is unknown, several risk factors may contribute to the onset and progression of IPF, such as aging, air pollution, smoking habits, microbial infections, and occupational exposure [102,105].
Several experimental studies have reported evidence that altered lung and systemic levels of zinc and zinc-regulatory molecules are associated with pulmonary fibrosis [28,29,30,97,106,107,108]. Zinc-deficiency exacerbated ventilation-induced lung damage in mice and rats [28,29]. Furthermore, Zhang et al. demonstrated that mice fed a zinc-deficient diet have increased oxidative stress and inflammation, a reduced activity of antioxidant enzymes, and a subsequently induced fibrosis in the lung compared to mice fed a normal diet [30]. A very recent study of single-cell RNA-seq analysis of epithelial cells collected from patients with IPF and aged injured mice identified a zinc–metabolic defect of alveolar progenitor cells (AEC2). A significant decrease in levels of the specific zinc transporter ZIP8 was shown in the AEC2. The authors also showed that AEC2-specific deletion of Zip8 in mice and the feeding of a low-zinc-content diet to mice leads to exacerbated bleomycin-induced lung fibrosis. These results suggest that zinc metabolism and the zinc transporter ZIP8 play critical roles in regulating alveolar progenitor renewal. During aging and in IPF, the zinc transporter ZIP8 of AEC2s is reduced, leading to impaired alveolar repair and causing pulmonary fibrosis.
Moreover, zinc in the particulate matter of atmospheric dust samples and zinc salts has been shown to be toxic in the lung of mice [97]. Importantly, this study revealed that after four weeks of zinc salts administration in mice, the mice showed significant induction of inflammation and fibrosis in the lung [97], suggesting that inhaling particulates that contain a high soluble metal content, like zinc, may have a crucial effect on the pulmonary cells. In addition, zinc oxide nanoparticles (ZnoNP) are widely used in different commercial products, such as food additives, personal hygiene products, paints, cosmetics, and textiles. ZnoNP prompted the proliferation of airway epithelial cells and pulmonary fibrosis in mice [108]. Another study by Wang et al. demonstrated that compared with controls, ZnoNP-exposed mice showed significantly reduced body weight and increased total protein, hydroxyproline content, and total cell numbers in their bronchoalveolar lavage fluid and malondialdehyde and nitric oxide levels in the lung [109]. The authors also found hyperplastic changes and inflammation in the lungs of the ZnoNP-exposed mice.
Furthermore, He et al. uncovered a novel mechanism of Cu, Zn-SOD-mediated, and Th2-independent M2 macrophage polarization and provided a potential therapeutic target for attenuating the promoted development of lung fibrosis [106]. These findings suggest that both zinc deficiency and toxicity can cause lung damage. Zinc levels in the body need to be tightly regulated to maintain homeostasis and prevent the occurrence and progression of diseases like pulmonary fibrosis.

3.5. ARDS

ARDS is a serious, life-threatening lung disease associated with various etiologies either through indirect or direct lung insults and is characterized by moderate-to-severe hypoxemia, abnormal respiratory system compliance, and acute pulmonary infiltrates. Clinical symptoms include shortness of breath, breathing difficulties, dizziness, and confusion. Previous studies showed significant down-regulation of zinc levels in the plasma of patients with ARDS compared to healthy controls [28,110]. Gonçalves et al. analyzed serum zinc levels of 269 critically ill patients infected by severe acute respiratory syndrome coronavirus 2 and correlated with its association with ARDS. The authors found that low serum levels and a high prevalence of low-serum zinc levels were associated with severe ARDS [31]. Zinc deficiency in alveolar macrophages and lung epitheliums can decrease lung barrier function, leading to respiratory distress syndrome [32]. In the lung of a murine sepsis model, zinc deficiency increased lung and other organ damage [33]. Gomez et al. found that lipid concentration, especially the phospholipids, is altered in the lungs of rats fed a zinc-deficient diet [111].
In contrast, inhalation of zinc fumes has been shown to cause ARDS in mice. However, the underlying mechanism(s) of how zinc deficiency induces lung injury in the context of ARDS is unclear. Future studies are needed to explore the potential mechanisms of how altered zinc homeostasis causes ARDS and whether dysregulation of zinc could be therapeutically targeted in the disease.

3.6. PH

PH is a complex vascular disease characterized by abnormally high pressure in the blood vessels that affects the arteries in the lungs and the right side of the heart. Based on their causes, there are five different groups of PH: pulmonary arterial hypertension (PAH), PH due to left heart disease, PH due to chronic lung disease (e.g., COPD, interstitial lung disease, CF) and hypoxia, PH due to chronic blood clots in the lungs, and PH due to unknown causes. There is no cure for PH, but there are treatments available to help improve symptoms and slow the progression of the disease. Thus, there is an urgent unmet need to develop effective therapies to treat the disease.
Emerging evidence suggests that altered zinc and zinc transporter protein levels are associated with the occurrence and progression of PH [34,35,36,37]. A previous study by Zhao et al. identified that plasmalemma zinc transporter ZIP12, encoded by the gene Slc39a12, plays a critical role in hypoxia-induced pulmonary vascular remodeling [34]. ZIP12 expression was shown to be up-regulated in remodeled pulmonary vascular tissues of humans, rats, and cows susceptible to hypoxia-induced PH. A significant up-regulation of ZIP12 was also found in the lung of idiopathic PAH patients and monocrotaline-treated rats, another well-established PH animal model. Further in vitro studies showed that siRNA-mediated inhibition of ZIP12 diminished hypoxia-induced increases in intracellular labile zinc contents and proliferation of pulmonary arterial smooth muscle cells (PASMC). Zip12 genetic disruption inhibited hypoxia-induced PH in rats, as evidenced by a significant decrease in pulmonary arterial pressure, right heart hypertrophy, and muscularization. While the results of these studies point to ZIP12 having a fundamental role in regulating pulmonary vascular homeostasis and remodeling under hypoxic conditions, the underlying mechanisms of how ZIP12 affects hypoxic responses remain unclear. Another study by Xiao et al. demonstrated that increased intracellular labile zinc, possibly from ZIP12, was linked with reduced phosphatases, increased transcription factor CREB-mediated activity, and PASMC proliferation [35]. Zhu et al. showed that ZIP12 contributed to phenotypic switching in hypoxia-induced PASMCs and promoted PH. The authors proposed that the HIF-1/ZIP12/pERK-signaling axis could facilitate hypoxia-induced phenotypic switching in PASMCs [36]. A very recent study showed that aberrant levels of zinc homeostasis (ZIP12, MT3), S1P signaling (S1PRs, SPNS2), and vascular remodeling (αSMA, FI, RVSP) are associated with each other in the monocrotaline-induced PH rat model, and BMPR2-targeted therapy might alleviate this condition [37]. Together, these findings suggest a new therapeutic avenue for preventing or treating PH by inhibiting ZIP12 and suppressing excursions of intracellular free zinc.

4. Conclusions and Future Roadmap

Emerging evidence has shown that zinc is critical for the homeostasis of multiple organs and systems at the cellular, molecular, and systemic levels. Zinc dyshomeostasis has been linked with several lung diseases, such as asthma, COPD, IPF, and CF. Currently, it largely remains unclear how altered zinc and zinc regulatory molecules contribute to the development and progression of lung disease. There are still several questions about zinc dyshomeostasis and lung diseases. Is a low zinc level a major or minor contributing factor in the development and progression of lung diseases? Are low zinc levels the cause or the result of lung diseases? Could zinc supplementation be effective as a treatment for each of these disorders if zinc insufficiency is the cause (Table 2)? Or would alternative measures, such as controlling the expression of zinc transporters and developing more targeted therapies, be required to restore zinc homeostasis and improve disease outcomes? More research on zinc homeostasis is needed to answer these questions, which could pave the way for new therapeutics targeting zinc homeostasis in lung inflammatory disorders.

Author Contributions

X.L.: Conceptualization, writing—original draft, revision; M.K.A., K.D.: Manuscript discussion, review, editing and revision; R.X.: Conceptualization, writing—review and editing, funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work is funded by the general project of Liaoning Natural Science Foundation, China (2021-MS-202).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Read, S.A.; Obeid, S.; Ahlenstiel, C.; Ahlenstiel, G. The Role of Zinc in Antiviral Immunity. Adv. Nutr. Int. Rev. J. 2019, 10, 696–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Wang, M.; Phadke, M.; Packard, D.; Yadav, D.; Gorelick, F. Zinc: Roles in pancreatic physiology and disease. Pancreatology 2020, 20, 1413–1420. [Google Scholar] [CrossRef] [PubMed]
  3. Skalny, A.V.; Aschner, M.; Tinkov, A.A. Zinc. Adv. Food. Nutr. Res. 2021, 96, 251–310. [Google Scholar] [PubMed]
  4. Hara, T.; Takeda, T.-A.; Takagishi, T.; Fukue, K.; Kambe, T.; Fukada, T. Physiological roles of zinc transporters: Molecular and genetic importance in zinc homeostasis. J. Physiol. Sci. 2017, 67, 283–301. [Google Scholar] [CrossRef]
  5. Hojyo, S.; Fukada, T. Zinc transporters and signaling in physiology and pathogenesis. Arch. Biochem. Biophys. 2016, 611, 43–50. [Google Scholar] [CrossRef]
  6. Suzuki, M.; Suzuki, T.; Watanabe, M.; Hatakeyama, S.; Kimura, S.; Nakazono, A.; Honma, A.; Nakamaru, Y.; Vreugde, S.; Homma, A. Role of intracellular zinc in molecular and cellular function in allergic inflammatory diseases. Allergol. Int. 2020, 70, 190–200. [Google Scholar] [CrossRef]
  7. Valera, P.; Zavattari, P.; Sanna, A.; Pretti, S.; Marcello, A.; Mannu, C.; Targhetta, C.; Bruno, G.; Songini, M. Zinc and Other Metals Deficiencies and Risk of Type 1 Diabetes: An Ecological Study in the High Risk Sardinia Island. PLoS ONE 2015, 10, e0141262. [Google Scholar] [CrossRef] [Green Version]
  8. Wessels, I.; Maywald, M.; Rink, L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017, 9, 1286. [Google Scholar] [CrossRef] [Green Version]
  9. Sanna, A.; Firinu, D.; Zavattari, P.; Valera, P. Zinc Status and Autoimmunity: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 68. [Google Scholar] [CrossRef] [Green Version]
  10. Thambiayya, K.; Kaynar, A.M.; Croix, C.M.S.; Pitt, B.R. Functional Role of Intracellular Labile Zinc in Pulmonary Endothelium. Pulm. Circ. 2012, 2, 443–451. [Google Scholar] [CrossRef] [Green Version]
  11. Pal, A.; Squitti, R.; Picozza, M.; Pawar, A.; Rongioletti, M.; Dutta, A.K.; Sahoo, S.; Goswami, K.; Sharma, P.; Prasad, R. Zinc and COVID-19: Basis of Current Clinical Trials. Biol. Trace Element Res. 2020, 199, 2882–2892. [Google Scholar] [CrossRef] [PubMed]
  12. Butters, D.; Whitehouse, M. COVID-19 and nutriceutical therapies, especially using zinc to supplement antimicrobials. Inflammopharmacology 2020, 29, 101–105. [Google Scholar] [CrossRef] [PubMed]
  13. Joachimiak, M.P. Zinc against COVID-19? Symptom surveillance and deficiency risk groups. PLOS Neglected Trop. Dis. 2021, 15, e0008895. [Google Scholar] [CrossRef]
  14. Kumar, A.; Kubota, Y.; Chernov, M.; Kasuya, H. Potential role of zinc supplementation in prophylaxis and treatment of COVID-19. Med. Hypotheses 2020, 144, 109848. [Google Scholar] [CrossRef]
  15. Wessels, I.; Rolles, B.; Rink, L. The Potential Impact of Zinc Supplementation on COVID-19 Pathogenesis. Front. Immunol. 2020, 11, 1712. [Google Scholar] [CrossRef] [PubMed]
  16. Skalny, A.V.; Rink, L.; Ajsuvakova, O.P.; Aschner, M.; Gritsenko, V.A.; Alekseenko, S.I.; Svistunov, A.A.; Petrakis, D.; Spandidos, D.A.; Aaseth, J.; et al. Zinc and respiratory tract infections: Perspectives for COVID-19 (Review). Int. J. Mol. Med. 2020, 46, 17–26. [Google Scholar] [CrossRef] [Green Version]
  17. Hunter, J.; Arentz, S.; Goldenberg, J.; Yang, G.; Beardsley, J.; Myers, S.P.; Mertz, D.; Leeder, S. Zinc for the prevention or treatment of acute viral respiratory tract infections in adults: A rapid systematic review and meta-analysis of randomised controlled trials. BMJ Open 2021, 11, e047474. [Google Scholar] [CrossRef]
  18. Mohamed, N.A.; Rushdy, M.; Abdel-Rehim, A.S. The immunomodulatory role of zinc in asthmatic patients. Cytokine 2018, 110, 301–305. [Google Scholar] [CrossRef]
  19. Truong-Tran, A.Q.; Ruffin, R.E.; Foster, P.S.; Koskinen, A.M.; Coyle, P.; Philcox, J.C.; Rofe, A.M.; Zalewski, P.D. Altered Zinc Homeostasis and Caspase-3 Activity in Murine Allergic Airway Inflammation. Am. J. Respir. Cell Mol. Biol. 2002, 27, 286–296. [Google Scholar] [CrossRef]
  20. Rerksuppaphol, S.; Rerksuppaphol, L. Zinc Supplementation in Children with Asthma Exacerbation. Pediatr. Rep. 2016, 8, 63–67. [Google Scholar] [CrossRef] [Green Version]
  21. Knoell, D.L.; Smith, D.A.; Sapkota, M.; Heires, A.J.; Hanson, C.K.; Smith, L.M.; Poole, J.A.; Wyatt, T.A.; Romberger, D.J. Insufficient zinc intake enhances lung inflammation in response to agricultural organic dust exposure. J. Nutr. Biochem. 2019, 70, 56–64. [Google Scholar] [CrossRef] [PubMed]
  22. Knoell, D.L.; Smith, D.; Bao, S.; Sapkota, M.; Wyatt, T.A.; Zweier, J.L.; Flury, J.; Borchers, M.T.; Knutson, M. Imbalance in zinc homeostasis enhances lung Tissue Loss following cigarette smoke exposure. J. Trace Elements Med. Biol. 2020, 60, 126483. [Google Scholar] [CrossRef] [PubMed]
  23. Hamon, R.; Homan, C.; Tran, H.B.; Mukaro, V.; Lester, S.E.; Roscioli, E.; Bosco, M.D.; Murgia, C.M.; Ackland, M.L.; Jersmann, H.P.; et al. Zinc and Zinc Transporters in Macrophages and Their Roles in Efferocytosis in COPD. PLoS ONE 2014, 9, e110056. [Google Scholar] [CrossRef] [Green Version]
  24. Roscioli, E.; Tran, H.B.; Jersmann, H.; Nguyen, P.T.; Hopkins, E.; Lester, S.E.; Farrow, N.; Zalewski, P.D.; Reynolds, P.N.; Hodge, S. The uncoupling of autophagy and zinc homeostasis in airway epithelial cells as a fundamental contributor to COPD. Am. J. Physiol. Cell. Mol. Physiol. 2017, 313, L453–L465. [Google Scholar] [CrossRef] [PubMed]
  25. Damphousse, V.; Mailhot, M.; Berthiaume, Y.; Rabasa-Lhoret, R.; Mailhot, G. Plasma zinc in adults with cystic fibrosis: Correlations with clinical outcomes. J. Trace Elements Med. Biol. 2014, 28, 60–64. [Google Scholar] [CrossRef] [PubMed]
  26. Kamei, S.; Fujikawa, H.; Nohara, H.; Ueno-Shuto, K.; Maruta, K.; Nakashima, R.; Kawakami, T.; Matsumoto, C.; Sakaguchi, Y.; Ono, T.; et al. Zinc Deficiency via a Splice Switch in Zinc Importer ZIP2/SLC39A2 Causes Cystic Fibrosis-Associated MUC5AC Hypersecretion in Airway Epithelial Cells. EBioMedicine 2018, 27, 304–316. [Google Scholar] [CrossRef] [Green Version]
  27. Zsembery, A.; Fortenberry, J.A.; Liang, L.; Bebok, Z.; Tucker, T.A.; Boyce, A.T.; Braunstein, G.M.; Welty, E.; Bell, P.D.; Sorscher, E.J.; et al. Extracellular Zinc and ATP Restore Chloride Secretion across Cystic Fibrosis Airway Epithelia by Triggering Calcium Entry. J. Biol. Chem. 2004, 279, 10720–10729. [Google Scholar] [CrossRef] [Green Version]
  28. Boudreault, F.; Pinilla-Vera, M.; Englert, J.A.; Kho, A.T.; Isabelle, C.; Arciniegas, A.J.; Barragan-Bradford, D.; Quintana, C.; Amador-Munoz, D.; Guan, J.; et al. Zinc deficiency primes the lung for ventilator-induced injury. JCI Insight 2017, 2, e86507. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, X.; Bian, J.; Ge, Y. Zinc-deficient diet aggravates ventilation-induced lung injury in rats. J. Biomed. Res. 2012, 26, 59–65. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, Q.; Xue, Y.; Fu, Y.; Bao, B.; Guo, M.-Y. Zinc Deficiency Aggravates Oxidative Stress Leading to Inflammation and Fibrosis in Lung of Mice. Biol. Trace Element Res. 2021, 1–13. [Google Scholar] [CrossRef]
  31. Goncalves, T.J.M.; Gonçalves, S.E.A.B.; Guarnieri, A.; Risegato, R.C.; Guimarães, M.P.; de Freitas, D.C.; Razuk-Filho, A.; Junior, P.B.B.; Parrillo, E.F. Association Between Low Zinc Levels and Severity of Acute Respiratory Distress Syndrome by New Coronavirus SARS-CoV-2. Nutr. Clin. Pract. 2021, 36, 186–191. [Google Scholar] [CrossRef] [PubMed]
  32. Skalny, A.V.; Skalnaya, M.G.; Grabeklis, A.R.; Skalnaya, A.A.; Tinkov, A.A. Zinc deficiency as a mediator of toxic effects of alcohol abuse. Eur. J. Nutr. 2017, 57, 2313–2322. [Google Scholar] [CrossRef] [PubMed]
  33. Knoell, D.L.; Julian, M.W.; Bao, S.; Besecker, B.; Macre, J.E.; Leikauf, G.; DiSilvestro, R.A.; Crouser, E.D. Zinc deficiency increases organ damage and mortality in a murine model of polymicrobial sepsis*. Crit. Care Med. 2009, 37, 1380–1388. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, L.; Oliver, E.; Maratou, K.; Atanur, S.S.; Dubois, O.D.; Cotroneo, E.; Chen, C.-N.; Wang, L.; Arce, C.; Chabosseau, P.L.; et al. The zinc transporter ZIP12 regulates the pulmonary vascular response to chronic hypoxia. Nature 2015, 524, 356–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Xiao, G.; Lian, G.; Wang, T.; Chen, W.; Zhuang, W.; Luo, L.; Wang, H.; Xie, L. Zinc-mediated activation of CREB pathway in proliferation of pulmonary artery smooth muscle cells in pulmonary hypertension. Cell Commun. Signal. 2021, 19, 1–16. [Google Scholar] [CrossRef]
  36. Zhu, T.; Wang, X.; Zheng, Z.; Quan, J.; Liu, Y.; Wang, Y.; Liu, T.; Liu, X.; Wang, M.; Zhang, Z. ZIP12 Contributes to Hypoxic Pulmonary Hypertension by Driving Phenotypic Switching of Pulmonary Artery Smooth Muscle Cells. J. Cardiovasc. Pharmacol. 2021, 79, 235–243. [Google Scholar] [CrossRef]
  37. Tran, H.B.; Maiolo, S.; Harper, R.; Zalewski, P.D.; Reynolds, P.N.; Hodge, S. Dysregulated zinc and sphingosine-1-phosphate signaling in pulmonary hypertension: Potential effects by targeting of bone morphogenetic protein receptor type 2 in pulmonary microvessels. Cell Biol. Int. 2021, 45, 2368–2379. [Google Scholar] [CrossRef]
  38. Truong-Tran, A.Q.; Carter, J.; Ruffin, R.; Zalewski, P.D. New insights into the role of zinc in the respiratory epithelium. Immunol. Cell Biol. 2001, 79, 170–177. [Google Scholar] [CrossRef]
  39. Bao, S.; Knoell, D.L. Zinc modulates cytokine-induced lung epithelial cell barrier permeability. Am. J. Physiol. Cell. Mol. Physiol. 2006, 291, L1132–L1141. [Google Scholar] [CrossRef] [Green Version]
  40. Stafford, S.L.; Bokil, N.J.; Achard, M.E.S.; Kapetanovic, R.; Schembri, M.A.; McEwan, A.G.; Sweet, M.J. Metal ions in macrophage antimicrobial pathways: Emerging roles for zinc and copper. Biosci. Rep. 2013, 33, e00049. [Google Scholar] [CrossRef]
  41. Liu, M.J.; Bao, S.; Gálvez-Peralta, M.; Pyle, C.J.; Rudawsky, A.C.; Pavlovicz, R.E.; Killilea, D.W.; Li, C.; Nebert, D.W.; Wewers, M.D.; et al. ZIP8 regulates host defense through zinc-mediated inhibition of NF-κB. Cell Rep. 2013, 3, 386–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Johnson, J.K.; Harris, F.L.; Ping, X.-D.; Gauthier, T.W.; Brown, L.A.S. Role of zinc insufficiency in fetal alveolar macrophage dysfunction and RSV exacerbation associated with fetal ethanol exposure. Alcohol 2018, 80, 5–16. [Google Scholar] [CrossRef] [PubMed]
  43. Mehta, A.J.; Yeligar, S.M.; Elon, L.; Brown, L.A.; Guidot, D.M. Alcoholism Causes Alveolar Macrophage Zinc Deficiency and Immune Dysfunction. Am. J. Respir. Crit. Care Med. 2013, 188, 716–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ali, M.K.; Kim, R.Y.; Karim, R.; Mayall, J.R.; Martin, K.L.; Shahandeh, A.; Abbasian, F.; Starkey, M.R.; Loustaud-Ratti, V.; Johnstone, D.; et al. Role of iron in the pathogenesis of respiratory disease. Int. J. Biochem. Cell Biol. 2017, 88, 181–195. [Google Scholar] [CrossRef]
  45. 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]
  46. Ariaee, N.; Farid, R.; Shabestari, F.; Shabestari, M.; Azad, F.J. Trace Elements Status in Sera of Patients with Allergic Asthma. Rep. Biochem. Mol. Biol. 2016, 5, 20–25. [Google Scholar]
  47. Kuti, B.P.; Kuti, D.K.; Smith, O.S. Serum Zinc, Selenium and Total Antioxidant Contents of Nigerian Children with Asthma: Association with Disease Severity and Symptoms Control. J. Trop. Pediatr. 2019, 66, 395–402. [Google Scholar] [CrossRef]
  48. Chen, M.; Sun, Y.; Wu, Y. Lower circulating zinc and selenium levels are associated with an increased risk of asthma: Evidence from a meta-analysis. Public Health Nutr. 2019, 23, 1555–1562. [Google Scholar] [CrossRef]
  49. Andino, D.; Moy, J.; Gaynes, B.I. Serum vitamin A, zinc and visual function in children with moderate to severe persistent asthma. J. Asthma 2018, 56, 1198–1203. [Google Scholar] [CrossRef]
  50. Khanbabaee, G.; Omidian, A.; Imanzadeh, F.; Adibeshgh, F.; Ashayeripanah, M.; Rezaei, N. Serum level of zinc in asthmatic patients: A case–control study. Allergol. Immunopathol. 2014, 42, 19–21. [Google Scholar] [CrossRef]
  51. Vural, H.; Uzun, K.; Uz, E.; Koçyigit, A.; Çigli, A.; Akyol, Ö. Concentrations of copper, zinc and various elements in serum of patients with bronchial asthma. J. Trace Elements Med. Biol. 2000, 14, 88–91. [Google Scholar] [CrossRef]
  52. Kadrabová, J.; Mad’Arić, A.; Podivínsky, F.; Gazdík, F.; Ginter, E. Plasma Zinc, copper and copper/zinc ratio in intrinsic asthma. J. Trace Elements Med. Biol. 1996, 10, 50–53. [Google Scholar] [CrossRef]
  53. Urushidate, S.; Matsuzaka, M.; Okubo, N.; Iwasaki, H.; Hasebe, T.; Tsuya, R.; Iwane, K.; Inoue, R.; Yamai, K.; Danjo, K.; et al. Association between concentration of trace elements in serum and bronchial asthma among Japanese general population. J. Trace Elements Med. Biol. 2010, 24, 236–242. [Google Scholar] [CrossRef] [PubMed]
  54. Kocyigit, A.; Armutcu, F.; Gurel, A.; Ermis, B. Alterations in Plasma Essential Trace Elements Selenium, Manganese, Zinc, Copper, and Iron Concentrations and the Possible Role of these Elements on Oxidative Status in Patients with Childhood Asthma. Biol. Trace Element Res. 2004, 97, 31–42. [Google Scholar] [CrossRef]
  55. Picado, C.; Deulofeu, R.; Lleonart, R.; Agusti, M.; Mullol, J.; Torra, M.; Quintó, L. Dietary micronutrients/antioxidants and their relationship with bronchial asthma severity. Allergy 2001, 56, 43–49. [Google Scholar] [CrossRef] [PubMed]
  56. AbdulWahab, A.; Zeidan, A.; Avades, T.; Chandra, P.; Soliman, A. Serum Zinc Level in Asthmatic and Non-Asthmatic School Children. Children 2018, 5, 42. [Google Scholar] [CrossRef] [Green Version]
  57. Mao, S.; Wu, L.; Shi, W. Association between trace elements levels and asthma susceptibility. Respir. Med. 2018, 145, 110–119. [Google Scholar] [CrossRef]
  58. Gray, R.D.; Duncan, A.; Noble, D.; Imrie, M.; O’Reilly, D.S.J.; Innes, J.A.; Porteous, D.; Greening, A.P.; Boyd, A.C. Sputum Trace Metals Are Biomarkers of Inflammatory and Suppurative Lung Disease. Chest 2010, 137, 635–641. [Google Scholar] [CrossRef] [Green Version]
  59. Jayaram, L.; Chunilal, S.; Pickering, S.; Ruffin, R.E.; Zalewski, P.D. Sputum zinc concentration and clinical outcome in older asthmatics. Respirology 2011, 16, 459–466. [Google Scholar] [CrossRef]
  60. Carneiro, M.F.H.; Rhoden, C.R.; Amantéa, S.L.; Barbosa, F.; Jr, F.B. Low Concentrations of Selenium and Zinc in Nails are Associated with Childhood Asthma. Biol. Trace Element Res. 2011, 144, 244–252. [Google Scholar] [CrossRef]
  61. Yilmaz, E.A.; Ozmen, S.; Bostanci, I.; Misirlioglu, E.D.; Ertan, U. Erythrocyte zinc levels in children with bronchial asthma. Pediatr. Pulmonol. 2011, 46, 1189–1193. [Google Scholar] [CrossRef] [PubMed]
  62. Uysalol, M.; Uysalol, E.P.; Yilmaz, Y.; Parlakgul, G.; Ozden, T.A.; Ertem, H.V.; Omer, B.; Uzel, N. Serum level of vitamin D and trace elements in children with recurrent wheezing: A cross-sectional study. BMC Pediatr. 2014, 14, 270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ribeiro-Silva, R.D.C.; Fiaccone, R.L.; Barreto, M.L.; da Silva, L.A.; Santos, L.F.P.; Alcantara-Neves, N.M. The prevalence of wheezing and its association with serum zinc concentration in children and adolescents in Brazil. J. Trace Elements Med. Biol. 2014, 28, 293–297. [Google Scholar] [CrossRef] [PubMed]
  64. Razi, C.H.; Akelma, A.Z.; Akin, O.; Kocak, M.; Ozdemir, O.; Celik, A.; Kislal, F.M. Hair zinc and selenium levels in children with recurrent wheezing. Pediatr. Pulmonol. 2012, 47, 1185–1191. [Google Scholar] [CrossRef] [PubMed]
  65. Richter, M.; Bonneau, R.; Girard, M.-A.; Beaulieu, C.; Larivée, P. Zinc status modulates bronchopulmonary eosinophil infiltration in a murine model of allergic inflammation. Chest 2003, 123, 446S. [Google Scholar] [CrossRef]
  66. Beckhaus, A.A.; Garcia-Marcos, L.; Forno, E.; Pacheco-Gonzalez, R.M.; Celedon, J.C.; Castro-Rodriguez, J.A. Maternal nutrition during pregnancy and risk of asthma, wheeze, and atopic diseases during childhood: A systematic review and meta-analysis. Allergy 2015, 70, 1588–1604. [Google Scholar] [CrossRef]
  67. Huang, K.-L.; Lee, Y.-H.; Chen, H.-I.; Liao, H.-S.; Chiang, B.-L.; Cheng, T.-J. Zinc oxide nanoparticles induce eosinophilic airway inflammation in mice. J. Hazard. Mater. 2015, 297, 304–312. [Google Scholar] [CrossRef]
  68. Huang, K.-L.; Chang, H.-L.; Tsai, F.-M.; Lee, Y.-H.; Wang, C.-H.; Cheng, T.-J. The effect of the inhalation of and topical exposure to zinc oxide nanoparticles on airway inflammation in mice. Toxicol. Appl. Pharmacol. 2019, 384, 114787. [Google Scholar] [CrossRef]
  69. Murray, C.J.; Lopez, A.D. Alternative projections of mortality and disability by cause 1990–2020: Global Burden of Disease Study. Lancet 1997, 349, 1498–1504. [Google Scholar] [CrossRef]
  70. Wewers, M.E.; Bailey, W.C.; Carlsen, K.-H.; Eisner, M.D.; Folan, P.; Heath, J.; Klinnert, M.D.; Kovesi, T.; Pien, G.W.; Reichart, V.C.; et al. An Official American Thoracic Society Workshop Report: Tobacco Control Initiatives within the American Thoracic Society. Proc. Am. Thorac. Soc. 2010, 7, 1–7. [Google Scholar] [CrossRef]
  71. Eisner, M.D.; Anthonisen, N.; Coultas, D.; Künzli, N.; Perez-Padilla, R.; Postma, D.; Romieu, I.; Silverman, E.K.; Balmes, J.R. An Official American Thoracic Society Public Policy Statement: Novel Risk Factors and the Global Burden of Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2010, 182, 693–718. [Google Scholar] [CrossRef] [PubMed]
  72. Pauwels, R.A.; Löfdahl, C.-G.; Laitinen, L.A.; Schouten, J.P.; Postma, D.S.; Pride, N.B.; Ohlsson, S.V. Long-Term Treatment with Inhaled Budesonide in Persons with Mild Chronic Obstructive Pulmonary Disease Who Continue Smoking. New Engl. J. Med. 1999, 340, 1948–1953. [Google Scholar] [CrossRef] [PubMed]
  73. Lin, Y.-S.; Caffrey, J.L.; Chang, M.-H.; Dowling, N.; Lin, J.-W. Cigarette smoking, cadmium exposure, and zinc intake on obstructive lung disorder. Respir. Res. 2010, 11, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Raguso, C.A.; Luthy, C. Nutritional status in chronic obstructive pulmonary disease: Role of hypoxia. Nutrition 2011, 27, 138–143. [Google Scholar] [CrossRef]
  75. Herzog, R.; Cunningham-Rundles, S. Immunologic impact of nutrient depletion in chronic obstructive pulmonary disease. Curr Drug Targets 2011, 12, 489–500. [Google Scholar] [CrossRef]
  76. Karadag, F.; Cildag, O.; Altinisik, M.; Kozaci, L.D.; Kiter, G.; Altun, C. Trace elements as a component of oxidative stress in COPD. Respirology 2004, 9, 33–37. [Google Scholar] [CrossRef]
  77. Shevcova, V.I.; Zujkova, A.A.; Pashkov, A.N.; Kotova, J.A.; Shevcov, A.N. Verification of zinc role in pathophysiology of chronic obstructive pulmonary disease. Ter. Arkhiv 2018, 90, 33–37. [Google Scholar] [CrossRef]
  78. El-Attar, M.; Said, M.; El-Assal, G.; Sabry, N.; Omar, E.; Ashour, L. Serum trace element levels in COPD patient: The relation between trace element supplementation and period of mechanical ventilation in a randomized controlled trial. Respirology 2009, 14, 1180–1187. [Google Scholar] [CrossRef]
  79. Anetor, J.I.; Ajose, F.; Anetor, G.O.; Iyanda, A.A.; Babalola, B.B.; Adeniyi, F.A.A. High cadmium/zinc ratio in cigarette smokers: Potential implications as a biomarker of risk of prostate cancer. Niger. J. Physiol. Sci. 2008, 23, 41–49. [Google Scholar] [CrossRef] [Green Version]
  80. Kazi, T.G.; Wadhwa, S.K.; Afridi, H.I.; Kazi, N.; Kandhro, G.A.; Baig, J.A.; Shah, A.Q.; Kolachi, N.F.; Khan, S. Evaluation of cadmium and zinc in biological samples of tobacco and alcohol user male mouth cancer patients. Hum. Exp. Toxicol. 2010, 29, 221–230. [Google Scholar] [CrossRef]
  81. Kırkıl, G.; Muz, M.H.; Seçkin, D.; Şahin, K.; Küçük, O. Antioxidant effect of zinc picolinate in patients with chronic obstructive pulmonary disease. Respir. Med. 2008, 102, 840–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Macdonald, R.S. The Role of Zinc in Growth and Cell Proliferation. J. Nutr. 2000, 130, 1500S–1508S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Mahabir, S.; Spitz, M.R.; Barrera, S.L.; Beaver, S.H.; Etzel, C.; Forman, M.R. Dietary zinc, copper and selenium, and risk of lung cancer. Int. J. Cancer 2007, 120, 1108–1115. [Google Scholar] [CrossRef] [PubMed]
  84. Collins, P.F.; Stratton, R.J.; Elia, M. Nutritional support in chronic obstructive pulmonary disease: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2012, 95, 1385–1395. [Google Scholar] [CrossRef] [Green Version]
  85. Collins, P.; Elia, M.; Stratton, R.J. Nutritional support and functional capacity in chronic obstructive pulmonary disease: A systematic review and meta-analysis. Respirology 2013, 18, 616–629. [Google Scholar] [CrossRef]
  86. Bauer, S.E.; Lai, H.J.; McDonald, C.M.; Asfour, F.; Slaven, J.E.; Ren, C.L. Zinc status and growth in infants and young children with cystic fibrosis. Pediatr. Pulmonol. 2021, 56, 3768–3776. [Google Scholar] [CrossRef]
  87. Akanli, L.; Lowenthal, D.B.; Gjonaj, S.; Dozor, A.J. Plasma and red blood cell zinc in cystic fibrosis. Pediatr. Pulmonol. 2002, 35, 2–7. [Google Scholar] [CrossRef]
  88. Wahab, A.A.; Abu-Shahin, A.; Allangawi, M.; Rahman, M.; Chandra, P. 206 Serum zinc concentration in cystic fibrosis patients with CFTR I1234V mutation associated with pancreatic sufficiency. J. Cyst. Fibros. 2014, 13, S99. [Google Scholar] [CrossRef] [Green Version]
  89. Van Biervliet, S.; Van Biervliet, J.P.; Vande Velde, S.; Robberecht, E. Serum zinc concentrations in cystic fibrosis patients aged above 4 years: A cross-sectional evaluation. Biol. Trace Elem. Res. 2007, 119, 19–26. [Google Scholar] [CrossRef] [Green Version]
  90. Krebs, N.F.; Westcott, J.E.; Arnold, T.D.; Kluger, B.M.; Accurso, F.J.; Miller, L.V.; Hambidge, K.M. Abnormalities in Zinc Homeostasis in Young Infants with Cystic Fibrosis. Pediatr. Res. 2000, 48, 256–261. [Google Scholar] [CrossRef] [Green Version]
  91. Abdulhamid, I.; Beck, F.; Millard, S.; Chen, X.; Prasad, A. Effect of zinc supplementation on respiratory tract infections in children with cystic fibrosis. Pediatr. Pulmonol. 2008, 43, 281–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sharma, G.K.; Lodha, R.; Shastri, S.; Saini, S.; Kapil, A.; Singla, M.; Mukherjee, A.; Jat, K.R.; Kabra, M.; Kabra, S.K. Zinc Supplementation for One Year Among Children with Cystic Fibrosis Does Not Decrease Pulmonary Infection. Respir. Care 2015, 61, 78–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. McBride, K.; Slotnick, B.; Margolis, F.L. Does intranasal application of zinc sulfate produce anosmia in the mouse? An olfactometric and anatomical study. Chem. Sens. 2003, 28, 659–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Fuortes, L.; Schenck, D. Marked elevation of urinary zinc levels and pleural-friction rub in metal fume fever. Vet. Hum. Toxicol. 2000, 42, 164–165. [Google Scholar]
  95. Pagan, I.; Costa, D.L.; McGee, J.K.; Richards, J.H.; Dye, J.A.; Dykstra, M.J. Metals Mimic Airway Epithelial Injury Induced by in Vitro Exposure to Utah Valley Ambient Particulate Matter Extracts. J. Toxicol. Environ. Health Part A 2003, 66, 1087–1112. [Google Scholar] [CrossRef]
  96. Gavett, S.H.; Haykal-Coates, N.; Copeland, L.B.; Heinrich, J.; Gilmour, M.I. Metal composition of ambient PM2.5 influences severity of allergic airways disease in mice. Environ. Health Perspect. 2003, 111, 1471–1477. [Google Scholar] [CrossRef] [Green Version]
  97. Adamson, I.; Prieditis, H.; Hedgecock, C.; Vincent, R. Zinc Is the Toxic Factor in the Lung Response to an Atmospheric Particulate Sample. Toxicol. Appl. Pharmacol. 2000, 166, 111–119. [Google Scholar] [CrossRef]
  98. Noble, P.W.; Barkauskas, C.E.; Jiang, D. Pulmonary fibrosis: Patterns and perpetrators. J. Clin. Investig. 2012, 122, 2756–2762. [Google Scholar] [CrossRef] [Green Version]
  99. Spagnolo, P.; Kropski, J.A.; Jones, M.G.; Lee, J.S.; Rossi, G.; Karampitsakos, T.; Maher, T.M.; Tzouvelekis, A.; Ryerson, C.J. Idiopathic pulmonary fibrosis: Disease mechanisms and drug development. Pharmacol. Ther. 2020, 222, 107798. [Google Scholar] [CrossRef]
  100. Raghu, G.; Remy-Jardin, M.; Myers, J.L.; Richeldi, L.; Ryerson, C.J.; Lederer, D.J.; Behr, J.; Cottin, V.; Danoff, S.K.; Morell, F.; et al. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2018, 198, e44–e68. [Google Scholar] [CrossRef]
  101. Raghu, G.; Weycker, D.; Edelsberg, J.; Bradford, W.Z.; Oster, G. Incidence and Prevalence of Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2006, 174, 810–816. [Google Scholar] [CrossRef] [PubMed]
  102. Raghu, G.; Collard, H.R.; Egan, J.J.; Martinez, F.J.; Behr, J.; Brown, K.K.; Colby, T.V.; Cordier, J.-F.; Flaherty, K.R.; Lasky, J.A.; et al. An Official ATS/ERS/JRS/ALAT Statement: Idiopathic Pulmonary Fibrosis: Evidence-based Guidelines for Diagnosis and Management. Am. J. Respir. Crit. Care Med. 2011, 183, 788–824. [Google Scholar] [CrossRef] [PubMed]
  103. Lederer, D.J.; Martinez, F.J. Idiopathic pulmonary fibrosis. New Engl. J. Med. 2018, 378, 1811–1823. [Google Scholar] [CrossRef] [PubMed]
  104. Hutchinson, J.P.; Fogarty, A.; Hubbard, R.B.; McKeever, T. Global incidence and mortality of idiopathic pulmonary fibrosis: A systematic review. Eur. Respir. J. 2015, 46, 795–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Sauleda, J.; Núñez, B.; Sala, E.; Soriano, J.B. Idiopathic Pulmonary Fibrosis: Epidemiology, Natural History, Phenotypes. Med Sci. 2018, 6, 110. [Google Scholar] [CrossRef] [Green Version]
  106. He, C.; Ryan, A.J.; Murthy, S.; Carter, A.B. Accelerated Development of Pulmonary Fibrosis via Cu,Zn-superoxide Dismutase-induced Alternative Activation of Macrophages. J. Biol. Chem. 2013, 288, 20745–20757. [Google Scholar] [CrossRef] [Green Version]
  107. Biaggio, V.S.; Salvetti, N.R.; Chaca, M.V.P.; Valdez, S.R.; Ortega, H.H.; Gimenez, M.S.; Gomez, N.N. Alterations of the extracellular matrix of lung during zinc deficiency. Br. J. Nutr. 2011, 108, 62–70. [Google Scholar] [CrossRef] [Green Version]
  108. Cho, W.-S.; Duffin, R.; Howie, S.E.; Scotton, C.J.; Wallace, W.A.; MacNee, W.; Bradley, M.; Megson, I.L.; Donaldson, K. Progressive severe lung injury by zinc oxide nanoparticles; the role of Zn2+ dissolution inside lysosomes. Part. Fibre Toxicol. 2011, 8, 27. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, D.; Li, H.; Liu, Z.; Zhou, J.; Zhang, T. Acute toxicological effects of zinc oxide nanoparticles in mice after intratracheal instillation. Int. J. Occup. Environ. Health 2017, 23, 11–19. [Google Scholar] [CrossRef]
  110. Cander, B.; Dundar, Z.D.; Gul, M.; Girisgin, S. Prognostic value of serum zinc levels in critically ill patients. J. Crit. Care 2011, 26, 42–46. [Google Scholar] [CrossRef] [Green Version]
  111. Gomez, N.N.; Ojeda, M.S.; Gimenez, M.S. Lung lipid composition in zinc-deficient rats. Lipids 2002, 37, 291–296. [Google Scholar] [CrossRef] [PubMed]
  112. Ghaffari, J.; Khalilian, A.; Salehifar, E.; Khorasani, E.; Rezaii, M. Effect of zinc supplementation in children with asthma: A randomized, placebo-controlled trial in northern Islamic Republic of Iran. East. Mediterr. Health J. 2014, 20, 391–396. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Role of zinc dysregulation in lung disease.
Figure 1. Role of zinc dysregulation in lung disease.
Nutrients 14 02115 g001
Table 1. Role of zinc and zinc-regulatory molecules in lung disease.
Table 1. Role of zinc and zinc-regulatory molecules in lung disease.
DiseaseZinc StatusPrimary outcomeMechanismReference
AsthmaLow serum zinc levels Serum zinc levels correlated with total IgE levels and forced expiratory volume in the first second (FEV1). Zinc triggered IFN-γ and inhibited IL-10 production in PBMCs of asthmatics[18]
Low zinc dietMice fed with a low zinc diet had greater levels of airway hyper-responsiveness (AHR), eosinophilia, and mucus cell hyperplasia, increased active caspase-3 and apoptosis-[19]
Zinc supplementationZinc supplementations improved asthma symptoms in asthma patients.-[20]
COPDZinc deficient dietZinc-deficient mice showed more severe airway inflammation in response to repeated agriculture dust exposure.-[21]
Zinc deficient diet, ZIP8 KOMice fed a restricted zinc diet had significantly increased CS-induced emphysema and ROS formation in the lung; Zip8 KO depletion or overexpression showed worse lung damage in mice exposed to chronic CS exposure. -[22]
Low zinc levels in bronchoalveolar lavage fluid (BALF) of COPD patients.Zinc chelatorZinc levels in BALF positively correlated with alveolar macrophage efferocytosis. TPEN significantly decreased efferocytosis in macrophages. Zinc transporters ZIP1 and ZIP2 differently responded to zinc-deficiency. -[23]
-Uncoupling role of zinc trafficking and autophagy in airway epithelial cells -[24]
CFLow plasma zinc in adult CF patientsLow zinc level was linked with worse clinical outcomes. -[25]
Low zinc levels due to ZIP2 splicing switch Zinc deficiency contributed to CF-associated MUC5AC hypersecretion in airway epithelial cells. -[26]
Extracellular zinc supplementation Extracellular zinc and ATP restored impaired chloride secretion in CF airway epithelium. Through stimulating calcium-dependent chloride channels[27]
IPFLow zinc dietZinc deficiency exacerbated ventilation-induced lung damage in mice and rats. -[28,29]
Low zinc dietMice fed a zinc-deficient diet had increased oxidative stress and inflammation, reduced activity of antioxidant enzymes, and subsequently induced fibrosis in the lung -[30]
ZIP8 KO, low zinc dietAEC2-specific deletion of Zip8 in mice and mice fed a low zinc content diet has exacerbated bleomycin-induced lung fibrosis.--
ARDSLow serum zinc of ARDS patientslow serum levels and a high prevalence of low serum zinc levels were associated with severe ARDS -[31].
zinc deficiencyZinc deficiency in alveolar macrophages and lung epitheliums decreased lung barrier function, leading to ARDS -[32].
Zinc deficiencyzinc deficiency induced lung and other organ damage-[33]
Zinc fume inhalationZinc fume developed ARDS in mice-
PHZIP12 upregulation in lung of IPAH patients, animal modelsZIP12 knockdown diminished hypoxia-induced increases in intracellular labile Zinc contents and proliferation of PASMC. Zip12 KO inhibited hypoxia-induced PH in rats, as evidenced by a significant decrease in pulmonary arterial pressure, right heart hypertrophy, and muscularization. [34]
High intracellular zinc, ZIP12 upregulation Increased intracellular labile zinc, possibly from ZIP12, was linked with reduced phosphatases, increased transcription factor CREB-mediated activity, and PASMC proliferation [35]
ZIP12 upregulationZIP12 contributed to hypoxia induced PASMCs phenotypic switch and promoted PH. HIF-1/ZIP12/pERK signaling axis could facilitate hypoxia-induced phenotypic switching in PASMCs[36]
Altered zinc homeostasisAltered levels of zinc homeostasis (ZIP12, MT3), S1P signaling (S1PRs, SPNS2), and vascular remodeling (αSMA, FI, RVSP) were associated with each other in the monocrotaline-induced PH rat model-[37]
Table 2. Clinical trials that study zinc supplementation in lung disease.
Table 2. Clinical trials that study zinc supplementation in lung disease.
DiseaseClinicaltrial.gov IDStudy DesignParticipantsStudy DurationIntervention Nutrient with DosagePrimary Outcome Key FindingsRef
AsthmaTCTR20141212001Double blinded RCT 4212 monthszinc bis-glycinate (30 mg elemental zinc/day)Pediatric respiratory assessment measure (PRAM) PRAM score decreased at 24 and 48 h[20]
-Double-blind, randomized, placebo-controlled clinical trial 2848 weeksZinc supplements (50 mg/day)Zinc balance and asthma clinical symptomsSignificantly improved zinc levels; clinical symptoms such as cough, wheezing, and dyspnoea; and lung function parameters (FVC, FEV1 and FEV1/FVC.[112]
CFNCT00104494Randomized, Parallel Assignment308 weeksZinc acetate (20 mg/day)Zinc balance --
-Double blind placebo-controlled pilot study2612 monthsZinc (30 mg/day)Rate of respiratory tract infections, antibiotics use, plasma cytokinesReduced the number of days of oral antibiotics used to treat RTIs in children with CF[91]
CTRI/2011/12/002230Double-blind randomized placebo-controlled trial4012 monthsZinc tablets
(30 mg/day)
A reduction in the average days of systemic antibiotics Zinc supplementation did not reduce lung infection in children with CF[92]
COPD-Randomized controlled trial308 weeksZinc picolinate
(22 mg/day)
Oxidant stress, and pulmonary function Favorable effects on oxidant–antioxidant balance[81]
-Double blinded RCT120-Sodium (100 mg/day);
zinc (2 mg/day); and
manganese (0.4 mg/day)
Effect of trace elements (Na, Mg, Zn) supplementation on the period the COPD patients spend on mechanical ventilationThe nutrition supplementation significantly reduced the period the patients with COPD spent on the mechanical ventilation [78]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, X.; Ali, M.K.; Dua, K.; Xu, R. The Role of Zinc in the Pathogenesis of Lung Disease. Nutrients 2022, 14, 2115. https://doi.org/10.3390/nu14102115

AMA Style

Liu X, Ali MK, Dua K, Xu R. The Role of Zinc in the Pathogenesis of Lung Disease. Nutrients. 2022; 14(10):2115. https://doi.org/10.3390/nu14102115

Chicago/Turabian Style

Liu, Xiaoying, Md Khadem Ali, Kamal Dua, and Ran Xu. 2022. "The Role of Zinc in the Pathogenesis of Lung Disease" Nutrients 14, no. 10: 2115. https://doi.org/10.3390/nu14102115

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