Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1314 2023-09-19 11:17:29 |
2 Format correct Meta information modification 1314 2023-09-19 12:51:21 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Seo, Y.; Park, J.; Kim, J.; Lee, M. Impact of Smoking on the Antioxidant System. Encyclopedia. Available online: https://encyclopedia.pub/entry/49376 (accessed on 07 September 2024).
Seo Y, Park J, Kim J, Lee M. Impact of Smoking on the Antioxidant System. Encyclopedia. Available at: https://encyclopedia.pub/entry/49376. Accessed September 07, 2024.
Seo, Yoon-Seok, Jung-Min Park, Jae-Hyeong Kim, Moo-Yeol Lee. "Impact of Smoking on the Antioxidant System" Encyclopedia, https://encyclopedia.pub/entry/49376 (accessed September 07, 2024).
Seo, Y., Park, J., Kim, J., & Lee, M. (2023, September 19). Impact of Smoking on the Antioxidant System. In Encyclopedia. https://encyclopedia.pub/entry/49376
Seo, Yoon-Seok, et al. "Impact of Smoking on the Antioxidant System." Encyclopedia. Web. 19 September, 2023.
Impact of Smoking on the Antioxidant System
Edit

Smoking is recognized as a significant risk factor for numerous disorders, including cardiovascular diseases, respiratory conditions, and various forms of cancer. While the exact pathogenic mechanisms continue to be explored, the induction of oxidative stress via the production of excess reactive oxygen species (ROS) is widely accepted as a primary molecular event that predisposes individuals to these smoking-related ailments. Oxidative stress represents a physiological condition characterized by an imbalance between oxidative and antioxidative potentials.

smoking cigarette smoke reactive oxygen species oxidative stress biomarkers antioxidant system

1. Introduction

Smoking is widely recognized as a significant risk factor for numerous diseases, including cardiovascular conditions, respiratory illnesses, and various types of cancers [1]. The toxicological implications of smoking are attributed to a number of mechanisms, many of which involve intricate molecular events. Thus, despite our increasing understanding, the molecular mechanisms continue to be a focus of rigorous investigation. Oxidative stress is widely identified as one of the key molecular events mediating the pathogenesis of smoking-associated diseases [2][3].
Oxidative stress represents a physiological condition characterized by an imbalance between oxidative and antioxidative potentials [4]. Such imbalance typically involves an excessive or deregulated production of prooxidants and/or a dysfunction within the antioxidant system. Prooxidants, which incite oxidative stress, comprise reactive oxygen species (ROS), radicals, and other oxidizing agents. In contrast, the antioxidant system involves both enzymatic antioxidants like superoxide dismutase (SOD), catalase, and glutathione (GSH) peroxidase, and non-enzymatic antioxidants such as GSH, carotenoids, and vitamins C and E [5].
Prooxidants, typified by ROS and radicals, represent highly reactive molecules that function as oxidizing agents in redox reactions. ROS, the primary endogenous prooxidants, are derivatives of molecular oxygen (O2) exhibiting greater reactivity compared to O2 itself [6]. These include species such as superoxide (O2•−), hydrogen peroxide (H2O2), hypochlorous acid (HOCl), and peroxynitrite/peroxynitrous acid (ONOO/ONOOH). These are produced endogenously through sources like the mitochondrial electron transport chain (ETC) and enzymes including NADPH oxidase (NOX), xanthine oxidase, nitric oxide synthase (NOS), and cytochromes P450 [7][8][9][10][11]. Moreover, they are generated via chemical reactions that involve transition metals like Fe and Cu [12]. Exogenous factors, including air pollutants and radiation, can also induce or stimulate their production [12]. The genesis of oxidative stress is not solely due to excessive or unregulated ROS formation but also linked to the dysregulation of the antioxidant system. Decreased antioxidant capacity can exacerbate ROS production and make biomolecules more susceptible to damage from prooxidants [13]. It is pertinent to mention that ROS, traditionally deemed hazardous byproducts of physiological processes or xenobiotics, also serve as signaling molecules, regulating various redox signaling involved in physiological processes [14]. ROS play crucial roles in diverse cellular events, such as growth, proliferation, differentiation, and apoptosis, by modulating redox-sensitive signaling molecules [14]. Furthermore, excessive ROS can inflict direct and nonspecific damage to various biomolecules, including lipids, proteins, and nucleic acids [13].

2. ROS Formation or Oxidative Stress in Smokers

Elevated oxidative stress, manifested by an intensified production of ROS, is a hallmark characteristic of smokers [4][8][15][16]. Such an observation is primarily grounded in clinical studies showcasing altered profiles of oxidative stress biomarkers in individuals who smoke. The majority of these biomarkers originate from the oxidative modification of biomolecules, encompassing lipid peroxidation products like 4-hydroxynonenal (4-HNE) [17], malondialdehyde (MDA) [18], and 8-isoprostane [19]; protein byproducts altered through oxidation such as protein carbonyls, 3-nitrotyrosine [20], and oxidized α-1 antitrypsin [21][22]; oxidized nucleic acid metabolites, such as 8-hydroxy-2′-deoxyguanosine (8-OHdG) [23]; and the antioxidant levels, notably GSH [24][25]. These biomarkers have been identified and quantified in an array of biological specimens including urine, blood, epithelial lining fluid, sputum, and saliva.
These oxidative stress biomarkers exhibit a tendency toward elevated levels within the respiratory system, the primary site of exposure to cigarette smoke [26][27][28]. For instance, 8-OHdG levels in the lung were discovered to be significantly higher in smokers than nonsmokers, with the degree of this elevation proportional to the cigarette smoking volume [26]. MDA levels were found to be increased in lung tissue samples from patients diagnosed with lung cancer [29]. Additionally, amplified levels of 8-isoprostane and 3-nitrotyrosine were observed in saliva and bronchial mucosa, respectively, from both asymptomatic smokers and patients diagnosed with chronic obstructive pulmonary disease (COPD), a common smoking-related condition [30][31].
Apart from the primary exposure site, biomarkers in blood and urine have also been examined to evaluate systemic oxidative stress [32]. Notably, the levels of MDA [32], 8-isoprostane [33][34], 3-nitrotyrosine [35], carbonyl content [36], and oxidized α-1 antitrypsin [21][22] in blood plasma or serum were higher in smokers compared to nonsmokers. Correspondingly, elevated urinary levels of 8-isoprostane [35], MDA [37], and 8-OHdG [38][39] were reported in smokers, demonstrating a quantitative correlation between the number of cigarettes consumed daily and the increment of these biomarkers [39]. It is crucial to note that the biomarker levels in blood or urine are influenced by both excretion and formation. Chronic smoking is known to impair renal function, which could potentially elevate these biomarker levels due to reduced excretion, in addition to the increased formation [23]. Therefore, kidney function must be factored into the interpretation of these findings. For example, the 8-isoprostane/creatinine ratio presents an effective index for oxidative stress assessment [35]. Moreover, high levels of 4-HNE and 8-OHdG have been identified in placenta samples from smoking mothers [40], indicating an association between maternal smoking and intrauterine oxidative stress. Smoking induces oxidative stress not only locally within the respiratory system but also at a systemic level.
Studies have substantiated the detrimental impact of smoking on the antioxidant system, precipitating a reduction in Trolox equivalent antioxidant capacity in plasma, an index of antioxidant capacity [41][42]. The literature abounds with evidence illustrating the adverse effects of smoking on antioxidants including, but not limited to, vitamins A, C, and E, carotene, and soluble thiol pools such as GSH [43][44]. A majority of these investigations have reported a decrease in these antioxidants consequent to smoking. However, a subset of studies has documented negligible alterations or even augmentation in GSH levels, potentially attributable to a compensatory mechanism countering depletion or a rebound phenomenon [45][46]. Although complicate molecular mechanisms are implicated, Nrf2 appears to play crucial roles in this compensation by upregulating enzymes responsible for GSH biosynthesis and regeneration [47]. Additionally, smoking has been found to suppress or downregulate enzymatic antioxidant systems including SOD and catalase in erythrocytes and plasma [48][49], as well as extracellular SOD in serum [50].
Given that the bulk of these studies have focused on chronic smokers, the perturbations in these biomarkers may represent the fallout of extended exposure to CS, suggesting that CS-induced oxidative stress could be a sequel to prolonged incidents such as smoking-induced inflammation or metabolic disorders. Nonetheless, even transient exposure to CS has been sufficient to amplify biomarkers of oxidative stress. As an example, the consumption of a single cigarette led to an upsurge in 8-isoprostane and soluble NOX2-derived peptide, a marker for NOX2 activation, along with a decrease in NO bioavailability and vitamin E in serum within 30 min [51][52]. Correspondingly, 8-OHdG levels in peripheral leukocytes were noted to rise a mere 10 min after smoking two cigarettes [53]. Passive smoking too has been found to increase plasma 8-isoprostane levels in nonsmokers within 30 min [54]. The rapid escalation in these biomarkers suggests an immediate induction of ROS formation by CS, irrespective of the presence of pathological conditions [55]. This notion is bolstered by findings that demonstrate the restoration of biomarker levels following smoking cessation, even in the absence of symptom improvement in patients with diabetes, hypercholesterolemia, or hypertension [56].
Regrettably, few studies have successfully detected a direct increase in ROS in reasonable ways rather than simply observing oxidative stress markers in human subjects, presumably due to the inherent technical challenges associated with measuring ROS. A pioneering study detected chemiluminescence in the blood plasma of smokers, which promptly ceased following smoking cessation, leading researchers to postulate its origin in ROS such as singlet oxygen derived from CS [57]. Another investigation, which analyzed expired breath condensate, reported elevated levels of hydrogen peroxide in smokers compared to nonsmokers, indicative of CS-induced hydrogen peroxide formation in the airway epithelial lining fluid [58]. This collective body of evidence intimates a direct elevation of ROS as a result of smoking.

References

  1. U.S. Department of Health Services. Chapter 1. Introduction, Summary, and Conclusions. In The Health Consequences of Smoking: 50 Years of Progress. A Report of the Surgeon General; U.S. Department of Health and Human Services; Centers for Disease Control and Prevention; National Center for Chronic Disease Prevention and Health Promotion; Office on Smoking and Health: Atlanta, GA, USA, 2014; pp. 1–14.
  2. Boukhenouna, S.; Wilson, M.A.; Bahmed, K.; Kosmider, B. Reactive oxygen species in chronic obstructive pulmonary disease. Oxid. Med. Cell. Longev. 2018, 2018, 5730395.
  3. Magder, S. Reactive oxygen species: Toxic molecules or spark of life? Crit. Care 2006, 10, 208.
  4. Ambrose, J.A.; Barua, R.S. The pathophysiology of cigarette smoking and cardiovascular disease: An update. J. Am. Coll. Cardiol. 2004, 43, 1731–1737.
  5. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19.
  6. Murphy, M.P.; Bayir, H.; Belousov, V.; Chang, C.J.; Davies, K.J.A.; Davies, M.J.; Dick, T.P.; Finkel, T.; Forman, H.J.; Janssen-Heininger, Y.; et al. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat. Metab. 2022, 4, 651–662.
  7. Li, X.; Fang, P.; Mai, J.; Choi, E.T.; Wang, H.; Yang, X.-F. Targeting mitochondrial reactive oxygen species as novel therapy for inflammatory diseases and cancers. J. Hematol. Oncol. 2013, 6, 19.
  8. Kim, M.; Han, C.H.; Lee, M.Y. NADPH oxidase and the cardiovascular toxicity associated with smoking. Toxicol. Res. 2014, 30, 149–157.
  9. Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine oxidoreductase-derived reactive species: Physiological and pathological effects. Oxid. Med. Cell. Longev. 2016, 2016, 3527579.
  10. Janaszak-Jasiecka, A.; Ploska, A.; Wieronska, J.M.; Dobrucki, L.W.; Kalinowski, L. Endothelial dysfunction due to eNOS uncoupling: Molecular mechanisms as potential therapeutic targets. Cell. Mol. Biol. Lett. 2023, 28, 21.
  11. Bou-Fakhredin, R.; Dia, B.; Ghadieh, H.E.; Rivella, S.; Cappellini, M.D.; Eid, A.A.; Taher, A.T. CYP450 mediates reactive oxygen species production in a mouse model of β-thalassemia through an increase in 20-HETE activity. Int. J. Mol. Sci. 2021, 22, 1106.
  12. Halliwell, B.; Gutteridge, J.M.C. Redox chemistry: The essentials. In Free Radicals in Biology and Medicine, 5th ed.; Oxford University Press: Oxford, UK, 2015; pp. 30–76.
  13. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462.
  14. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313.
  15. Grassi, D.; Desideri, G.; Ferri, L.; Aggio, A.; Tiberti, S.; Ferri, C. Oxidative stress and endothelial dysfunction: Say NO to cigarette smoking! Curr. Pharm. Des. 2010, 16, 2539–2550.
  16. Burke, A.; Fitzgerald, G.A. Oxidative stress and smoking-induced vascular injury. Prog. Cardiovasc. Dis. 2003, 46, 79–90.
  17. Breitzig, M.; Bhimineni, C.; Lockey, R.; Kolliputi, N. 4-Hydroxy-2-nonenal: A critical target in oxidative stress? Am. J. Physiol. Cell Physiol. 2016, 311, C537–C543.
  18. Nielsen, F.; Mikkelsen, B.B.; Nielsen, J.B.; Andersen, H.R.; Grandjean, P. Plasma malondialdehyde as biomarker for oxidative stress: Reference interval and effects of life-style factors. Clin. Chem. 1997, 43, 1209–1214.
  19. Montuschi, P.; Barnes, P.J.; Roberts, L.J., Jr. Isoprostanes: Markers and mediators of oxidative stress. FASEB J. 2004, 18, 1791–1800.
  20. Dalle-Donne, I.; Rossi, R.; Giustarini, D.; Milzani, A.; Colombo, R. Protein carbonyl groups as biomarkers of oxidative stress. Clin. Chim. Acta 2003, 329, 23–38.
  21. Carp, H.; Miller, F.; Hoidal, J.R.; Janoff, A. Potential mechanism of emphysema: Alpha 1-proteinase inhibitor recovered from lungs of cigarette smokers contains oxidized methionine and has decreased elastase inhibitory capacity. Proc. Natl. Acad. Sci. USA 1982, 79, 2041–2045.
  22. Topic, A.; Milovanovic, V.; Lazic, Z.; Ivosevic, A.; Radojkovic, D. Oxidized α1-antitrypsin as a potential biomarker associated with onset and severity of chronic obstructive pulmonary disease in adult population. COPD 2018, 15, 472–478.
  23. Campos, C.; Guzman, R.; Lopez-Fernandez, E.; Casado, A. Urinary biomarkers of oxidative/nitrosative stress in healthy smokers. Inhal. Toxicol. 2011, 23, 148–156.
  24. Ho, E.; Karimi Galougahi, K.; Liu, C.C.; Bhindi, R.; Figtree, G.A. Biological markers of oxidative stress: Applications to cardiovascular research and practice. Redox Biol. 2013, 1, 483–491.
  25. Rahman, I.; MacNee, W. Lung glutathione and oxidative stress: Implications in cigarette smoke-induced airway disease. Am. J. Physiol. 1999, 277, L1067–L1088.
  26. Asami, S.; Manabe, H.; Miyake, J.; Tsurudome, Y.; Hirano, T.; Yamaguchi, R.; Itoh, H.; Kasai, H. Cigarette smoking induces an increase in oxidative DNA damage, 8-hydroxydeoxyguanosine, in a central site of the human lung. Carcinogenesis 1997, 18, 1763–1766.
  27. Sundar, I.K.; Yao, H.; Kirkham, P.A.; Rahman, I. Smoking, oxidative/carbonyl stress, and regulation of redox signaling in lung inflammation. In Systems Biology of Free Radicals and Antioxidants; Laher, I., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 817–848.
  28. Kirkham, P.A.; Barnes, P.J. Oxidative stress in COPD. Chest 2013, 144, 266–273.
  29. Petruzzelli, S.; Hietanen, E.; Bartsch, H.; Camus, A.M.; Mussi, A.; Angeletti, C.A.; Saracci, R.; Giuntini, C. Pulmonary lipid peroxidation in cigarette smokers and lung cancer patients. Chest 1990, 98, 930–935.
  30. Kinnula, V.L.; Ilumets, H.; Myllarniemi, M.; Sovijarvi, A.; Rytila, P. 8-Isoprostane as a marker of oxidative stress in nonsymptomatic cigarette smokers and COPD. Eur. Respir. J. 2007, 29, 51–55.
  31. Ricciardolo, F.L.; Caramori, G.; Ito, K.; Capelli, A.; Brun, P.; Abatangelo, G.; Papi, A.; Chung, K.F.; Adcock, I.; Barnes, P.J.; et al. Nitrosative stress in the bronchial mucosa of severe chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2005, 116, 1028–1035.
  32. Block, G.; Dietrich, M.; Norkus, E.P.; Morrow, J.D.; Hudes, M.; Caan, B.; Packer, L. Factors associated with oxidative stress in human populations. Am. J. Epidemiol. 2002, 156, 274–285.
  33. Morrow, J.D.; Frei, B.; Longmire, A.W.; Gaziano, J.M.; Lynch, S.M.; Shyr, Y.; Strauss, W.E.; Oates, J.A.; Roberts, L.J., Jr. Increase in circulating products of lipid peroxidation (F2-isoprostanes) in smokers. Smoking as a cause of oxidative damage. N. Engl. J. Med. 1995, 332, 1198–1203.
  34. van ‘t Erve, T.J.; Lih, F.B.; Kadiiska, M.B.; Deterding, L.J.; Mason, R.P. Elevated plasma 8-iso-prostaglandin F2α levels in human smokers originate primarily from enzymatic instead of non-enzymatic lipid peroxidation. Free Radic. Biol. Med. 2018, 115, 105–112.
  35. Sakano, N.; Takahashi, N.; Wang, D.H.; Sauriasari, R.; Takemoto, K.; Kanbara, S.; Sato, Y.; Takigawa, T.; Takaki, J.; Ogino, K. Plasma 3-nitrotyrosine, urinary 8-isoprostane and 8-OHdG among healthy Japanese people. Free Radic. Res. 2009, 43, 183–192.
  36. Lee, B.M.; Lee, S.K.; Kim, H.S. Inhibition of oxidative DNA damage, 8-OHdG, and carbonyl contents in smokers treated with antioxidants (vitamin E, vitamin C, β-carotene and red ginseng). Cancer Lett. 1998, 132, 219–227.
  37. Klaunig, J.E.; Xu, Y.; Han, C.; Kamendulis, L.M.; Chen, J.; Heiser, C.; Gordon, M.S.; Mohler, E.R., 3rd. The effect of tea consumption on oxidative stress in smokers and nonsmokers. Proc. Soc. Exp. Biol. Med. 1999, 220, 249–254.
  38. Prieme, H.; Loft, S.; Klarlund, M.; Gronbaek, K.; Tonnesen, P.; Poulsen, H.E. Effect of smoking cessation on oxidative DNA modification estimated by 8-oxo-7,8-dihydro-2′-deoxyguanosine excretion. Carcinogenesis 1998, 19, 347–351.
  39. Reilly, M.; Delanty, N.; Lawson, J.A.; FitzGerald, G.A. Modulation of oxidant stress in vivo in chronic cigarette smokers. Circulation 1996, 94, 19–25.
  40. Sbrana, E.; Suter, M.A.; Abramovici, A.R.; Hawkins, H.K.; Moss, J.E.; Patterson, L.; Shope, C.; Aagaard-Tillery, K. Maternal tobacco use is associated with increased markers of oxidative stress in the placenta. Am. J. Obstet. Gynecol. 2011, 205, e1–e246.
  41. Rahman, I.; Morrison, D.; Donaldson, K.; MacNee, W. Systemic oxidative stress in asthma, COPD, and smokers. Am. J. Respir. Crit. Care Med. 1996, 154, 1055–1060.
  42. Petruzzelli, S.; Puntoni, R.; Mimotti, P.; Pulera, N.; Baliva, F.; Fornai, E.; Giuntini, C. Plasma 3-nitrotyrosine in cigarette smokers. Am. J. Respir. Crit. Care Med. 1997, 156, 1902–1907.
  43. Chow, C.K.; Thacker, R.R.; Changchit, C.; Bridges, R.B.; Rehm, S.R.; Humble, J.; Turbek, J. Lower levels of vitamin C and carotenes in plasma of cigarette smokers. J. Am. Coll. Nutr. 1986, 5, 305–312.
  44. Munro, L.H.; Burton, G.; Kelly, F.J. Plasma RRR-α-tocopherol concentrations are lower in smokers than in non-smokers after ingestion of a similar oral load of this antioxidant vitamin. Clin. Sci. 1997, 92, 87–93.
  45. Cantin, A.M.; Hanrahan, J.W.; Bilodeau, G.; Ellis, L.; Dupuis, A.; Liao, J.; Zielenski, J.; Durie, P. Cystic fibrosis transmembrane conductance regulator function is suppressed in cigarette smokers. Am. J. Respir. Crit. Care Med. 2006, 173, 1139–1144.
  46. Gould, N.S.; Min, E.; Huang, J.; Chu, H.W.; Good, J.; Martin, R.J.; Day, B.J. Glutathione depletion accelerates cigarette smoke-induced inflammation and airspace enlargement. Toxicol. Sci. 2015, 147, 466–474.
  47. Cantin, A.M. Cellular response to cigarette smoke and oxidants: Adapting to survive. Proc. Am. Thorac. Soc. 2010, 7, 368–375.
  48. Yildiz, L.; Kayaoglu, N.; Aksoy, H. The changes of superoxide dismutase, catalase and glutathione peroxidase activities in erythrocytes of active and passive smokers. Clin. Chem. Lab. Med. 2002, 40, 612–615.
  49. Tavilani, H.; Nadi, E.; Karimi, J.; Goodarzi, M.T. Oxidative stress in COPD patients, smokers, and non-smokers. Respir. Care 2012, 57, 2090–2094.
  50. Kim, S.H.; Kim, J.S.; Shin, H.S.; Keen, C.L. Influence of smoking on markers of oxidative stress and serum mineral concentrations in teenage girls in Korea. Nutrition 2003, 19, 240–243.
  51. Carnevale, R.; Sciarretta, S.; Violi, F.; Nocella, C.; Loffredo, L.; Perri, L.; Peruzzi, M.; Marullo, A.G.; De Falco, E.; Chimenti, I.; et al. Acute impact of tobacco versus electronic cigarette smoking on oxidative stress and vascular function. Chest 2016, 150, 606–612.
  52. Cave, A.C.; Brewer, A.C.; Narayanapanicker, A.; Ray, R.; Grieve, D.J.; Walker, S.; Shah, A.M. NADPH oxidases in cardiovascular health and disease. Antioxid. Redox Signal. 2006, 8, 691–728.
  53. Kiyosawa, H.; Suko, M.; Okudaira, H.; Murata, K.; Miyamoto, T.; Chung, M.H.; Kasai, H.; Nishimura, S. Cigarette smoking induces formation of 8-hydroxydeoxyguanosine, one of the oxidative DNA damages in human peripheral leukocytes. Free Radic. Res. Commun. 1990, 11, 23–27.
  54. Kato, T.; Inoue, T.; Morooka, T.; Yoshimoto, N.; Node, K. Short-term passive smoking causes endothelial dysfunction via oxidative stress in nonsmokers. Can. J. Physiol. Pharmacol. 2006, 84, 523–529.
  55. Luc, K.; Schramm-Luc, A.; Guzik, T.J.; Mikolajczyk, T.P. Oxidative stress and inflammatory markers in prediabetes and diabetes. J. Physiol. Pharmacol. 2019, 70, 809–824.
  56. Pilz, H.; Oguogho, A.; Chehne, F.; Lupattelli, G.; Palumbo, B.; Sinzinger, H. Quitting cigarette smoking results in a fast improvement of in vivo oxidation injury (determined via plasma, serum and urinary isoprostane). Thromb. Res. 2000, 99, 209–221.
  57. Yoda, B.; Goto, Y.; Sato, K.; Saeki, A.; Inaba, H. Ultra-weak chemiluminescence of smokers’ blood. Arch. Environ. Health 1985, 40, 148–150.
  58. Nowak, D.; Antczak, A.; Krol, M.; Pietras, T.; Shariati, B.; Bialasiewicz, P.; Jeczkowski, K.; Kula, P. Increased content of hydrogen peroxide in the expired breath of cigarette smokers. Eur. Respir. J. 1996, 9, 652–657.
More
Information
Subjects: Toxicology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 292
Revisions: 2 times (View History)
Update Date: 19 Sep 2023
1000/1000
ScholarVision Creations