#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Effects of selected heavy metals on the metabolism and healing processes of craniofacial bones


Authors: Bijowski Kamil 1;  Bortnik Piotr 1;  Kornowska Gabriela 2;  Dziewski Go Dawid 2;  Lewandowska Patrycja 2;  Czachorowski Antoni 2;  Borys Jan 1
Authors place of work: Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok/Klinika Chirurgii Szczękowo-Twarzowej i Plastycznej, Uniwersytet Medyczny w Białymstoku/Białystok, Poland 1;  Student science club at the Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok/Studenckie Koło Naukowe, Klinika Chirurgii Szczękowo-Twarzowej i Plastycznej, Uniwersytet Medyczny w Białymstoku/Białystok, Poland 2
Published in the journal: Clinical Osteology 2023; 28(4): 133-138
Category: Přehledové články

Summary

Introduction: The healing of craniofacial bones is a complex and multi-stage process that can be influenced by many factors of endogenous and exogenous origin. These factors include heavy metals, which play a significant role in the metabolism of the human body. Fractures of the craniofacial bones carry a particular risk, both because of their proximity to many important anatomical structures but also because of the function they represent for the beginning of two important systems: the digestive system and the respiratory system. It is therefore important to restore full function and normal bone metabolism as soon as possible. Objective: The aim of this study was to review the scientific literature on the effects of selected heavy metals: cadmium, zinc, lead, mercury, iron on the metabolism and healing processes of craniofacial bones. Material and methods: An analysis of the available sources shows that cadmium, zinc and lead have a negative impact on the physiological processes leading to skeletal fusion. In contrast, iron play a positive role in bone-forming processes. The effect of mercury on craniofacial bone metabolism is not yet fully understood. Summary: In summary, it can be concluded that heavy metals affect the healing processes and metabolism of craniofacial bones to varying degrees. The impact of these substances is not always negative. It should be borne in mind that it is extremely important to minimise the supply of some of these substances during the healing process directed at bone fusion.


Zdroje
  1. Mackenbach JP, Damhuis RA, Been JV. De gezondheidseffecten van roken. [The effects of smoking on health: growth of knowledge reveals even grimmer picture]. Ned Tijdschr Geneeskd 2017; 160: D869.
  2. De la Vega RE, Atasoy-Zeybek A, Panos JA et al. Gene therapy for bone healing: lessons learned and new approaches. Transl Res 2021; 236: 1–16. Available on DOI: <http://dx.doi.org/10.1016/j.trsl.2021.04.009>.
  3. Åkesson A, Barregard L, Bergdahl IA et al. Non-renal effects and the risk assessment of environmental cadmium exposure. Environ Health Perspect 2014; 122(5): 431–438. Available on DOI:<http://dx.doi.org/10.1289/ehp.1307110>.
  4. Feki-Tounsi M, Hamza-Chaffai A. Cadmium as a possible cause of bladder cancer: a review of accumulated evidence. Environ Sci Pollut Res Int 2014; 21(18): 10561–10573. Available on DOI: <http://dx.doi.org/10.1007/s11356–014–2970–0>.
  5. Satarug S, Baker JR, Urbenjapol S et al. A global perspective on cadmium pollution and toxicity in non-occuptionally exposed population. Toxicol Lett 2003; 137(1–2): 65–83. Available on DOI:<http://dx.doi.org/10.1016/s0378–4274(02)00381–8>.
  6. Satarug S, Moore MR. Adverse health effects of chronic exposure to low-level cadmium in foodstuffs and cigarette smoke. Environ Health Perspect 2004; 112(10): 1099–1103. Available on DOI: <http://dx.doi.org/10.1289/ehp.6751>.
  7. Graniel-Amador MA, Torres-Rodríguez HF, Jiménez-Andrade JM et al. Cadmium exposure negatively affects the microarchitecture of trabecular bone and decreases the density of a subset of sympathetic nerve fibers innervating the developing rat femur. Biometals 2021; 34(1): 87–96. Available on DOI: <http://dx.doi.org/10.1007/s10534–020–00265-x>.
  8. Qing Y, Yang J, Zhu Y et al. Dose-response evaluation of urinary cadmium and kidney injury biomarkers in Chinese residents and dietary limit standards. Environ Health 2021; 20(1): 75. Available on DOI: <http://dx.doi.org/10.1186/s12940–021–00760–9>.
  9. Wallin M, Barregard L, Sallsten G et al. Low-level cadmium exposure is associated with decreased cortical thickness, cortical area and trabecular bone volume fraction in elderly men: The MrOS Sweden study. Bone 2021; 143: 115768. Available on DOI: <http://dx.doi.org/10.1016/j.bone.2020.115768>.
  10. Ahmed MF, Mokhtar MB. Assessing Cadmium and Chromium Concentrations in Drinking Water to Predict Health Risk in Malaysia. Int J Environ Res Public Health 2020; 17(8): 2966. Available on DOI: <http://dx.doi.org/10.3390/ijerph17082966.
  11. Engström A, Michaëlsson K, Suwazono Y et al. Long-term cadmium exposure and the association with bone mineral density and fractures in a population-based study among women. J Bone Miner Res 2011; 26(3): 486–495. Available on DOI: <http://dx.doi.org/10.1002/jbmr.224>.
  12. Sughis M, Penders J, Haufroid V et al. Bone resorption and environmental exposure to cadmium in children: a cross-sectional study. Environ Health 2011; 10: 104. Available on DOI: <http://dx.doi.org/10.1186/1476–069X-10–104>.
  13. Tapiero H, Tew KD. Trace elements in human physiology and pathology: zinc and metallothioneins. Biomed Pharmacother 2003; 57(9): 399–411. Available on DOI: <http://dx.doi.org/10.1016/s0753–3322(03)00081–7>.
  14. Miggiano GA, Gagliardi L. Dieta, nutrizione e salute dell’osso. [Diet, nutrition and bone health]. Clin Ter 2005; 156(1–2): 47–56.
  15. Uchiyama S, Ishiyama K, Hashimoto K et al. Synergistic effect of beta-cryptoxanthin and zinc sulfate on the bone component in rat femoral tissues in vitro: the unique anabolic effect with zinc. Biol Pharm Bull 2005; 28(11): 2142–2145. Available on DOI: <http://dx.doi.org/10.1248/bpb.28.2142>.
  16. Hill T, Meunier N, Andriollo-Sanchez M et al. The relationship between the zinc nutritive status and biochemical markers of bone turnover in older European adults: the ZENITH study. Eur J Clin Nutr 2005; 59(Suppl 2): S73-S78. Available on DOI: <http://dx.doi.org/10.1038/sj.ejcn.1602303>.
  17. Hosea HJ, Taylor CG, Wood T et al. Zinc-deficient rats have more limited bone recovery during repletion than diet-restricted rats. Exp Biol Med (Maywood) 2004; 229(4): 303–311. Available on DOI: <http://dx.doi.org/10.1177/153537020422900404Z>.
  18. Huang T, Yan G, Guan M. Zinc Homeostasis in Bone: Zinc Transporters and Bone Diseases. Int J Mol Sci 2020; 21(4): 1236. Available on DOI: <http://dx.doi.org/10.3390/ijms21041236>.
  19. Sadighi A, Roshan MM, Moradi A et al. The effects of zinc supplementation on serum zinc, alkaline phosphatase activity and fracture healing of bones. Saudi Med J 2008; 29(9): 1276–1279. Erratum in Saudi Med J 2008; 29(12): 1836.
  20. Yan S, Liu Y, Tian X et al. Effect of extraneous zinc on calf intestinal alkaline phosphatase. J Protein Chem 2003; 22(4): 371–375. Available on DOI: <http://dx.doi.org/10.1023/a:1025394224669>.
  21. Begam H, Nandi SK, Chanda A et al. Effect of bone morphogenetic protein on Zn-HAp and Zn-HAp/collagen composite: A systematic in vivo study. Res Vet Sci 2017; 115: 1–9. Available on DOI: <http://dx.doi.org/10.1016/j.rvsc.2017.01.012>.
  22. Zhong Y, Li X, Hu DY et al. Control of Established Gingivitis and Dental Plaque Using a 1450 ppm Fluoride/Zinc-based Dentifrice: A Randomized Clinical Study. J Clin Dent 2015; 26(4): 104–108.
  23. Seyedmajidi SA, Seyedmajidi M, Moghadamnia A et al. Effect of zinc-deficient diet on oral tissues and periodontal indices in rats. Int J Mol Cell Med 2014; 3(2): 81–87.
  24. Li P, Zhang W, Dai J et al. Investigation of zinc copper alloys as potential materials for craniomaxillofacial osteosynthesis implants. Mater Sci Eng C Mater Biol Appl 2019; 103: 109826. Available on DOI: <http://dx.doi.org/10.1016/j.msec.2019.109826>. 
  25. Xia D, Yang F, Zheng Y et al. Research status of biodegradable metals designed for oral and maxillofacial applications: A review. Bioact Mater 2021; 6(11): 4186–4208. Available on DOI: <http://dx.doi.org/10.1016/j.bioactmat.2021.01.011>.
  26. Tokudome Y, Otsuka M. Possibility of alveolar bone promoting enhancement by using lipophilic and/or hydrophilic zinc related compounds in zinc-deficient osteoporosis rats. Biol Pharm Bull 2012; 35(9): 1496–1501. Available on DOI: <http://dx.doi.org/10.1248/bpb.b12–00218>.
  27. Guo H, Xia D, Zheng Y et al. A pure zinc membrane with degradability and osteogenesis promotion for guided bone regeneration: In vitro and in vivo studies. Acta Biomater 2020; 106: 396–409. Available on DOI: <http://dx.doi.org/10.1016/j.actbio.2020.02.024>. 
  28. Chou J, Komuro M, Hao J et al. Bioresorbable zinc hydroxyapatite guided bone regeneration membrane for bone regeneration. Clin Oral Implants Res 2016; 27(3): 354–360. Available on DOI: <http://dx.doi.org/10.1111/clr.12520>. 
  29. Wilk A, Kalisińska E, Różański J et al. Kadm, ołów i rtęć w nerkach człowieka. Medycyna Środowiskowa – Environmental Medicine 2013; 16(1): 75–81.
  30. Ciria-Recasens M, Blanch-Rubió J, Coll-Batet M et al. Comparison of the effects of ossein-hydroxyapatite complex and calcium carbonate on bone metabolism in women with senile osteoporosis: a randomized, open-label, parallel-group, controlled, prospective study. Clin Drug Investig 2011; 31(12): 817–824. Available on DOI: <http://dx.doi.org/10.1007/BF03256920>. 
  31. Álvarez-Lloret P, Benavides-Reyes C, Lee CM et al. Chronic Lead Exposure Alters Mineral Properties in Alveolar Bone. Minerals 2021; 11(6): 642. Available on DOI: <https://doi.org/10.3390/min11060642>. 
  32. Han DH, Lee HJ, Lim S. Smoking induced heavy metals and periodontitis: findings from the Korea National Health and Nutrition Examination Surveys 2008–2010. J Clin Periodontol 2013; 40(9): 850–858. Available on DOI: <http://dx.doi.org/10.1111/jcpe.12133>. 
  33. Kim Y, Lee BK. Association between blood lead and mercury levels and periodontitis in the Korean general population: analysis of the 2008–2009 Korean National Health and Nutrition Examination Survey data. Int Arch Occup Environ Health 2013; 86(5): 607–613. Available on DOI: <http://dx.doi.org/10.1007/s00420–012–0796-y>. 
  34. Ye X, Qian H, Xu P et al. Nephrotoxicity, neurotoxicity, and mercury exposure among children with and without dental amalgam fillings. Int J Hyg Environ Health 2009; 212(4): 378-386. Available on DOI: <http://doi: 10.1016/j.ijheh.2008.09.004>.
  35. Łanocha N, Kalisińska E, Kosik-Bogacka DI et al. Concentrations of trace elements in bones of the hip joint from patients aft er hip replacement surgery. J Trace Elem Med Biol 2012; 26(1): 20–25. Available on DOI: <http://dx.doi.org/10.1016/j.jtemb.2011.11.006>. 
  36. Nunes PBO, Ferreira MK, Ribeiro Frazão D et al. Effects of inorganic mercury exposure in the alveolar bone of rats: an approach of qualitative and morphological aspects. Peer J 2022; 10: e12573. Available on DOI: <http://dx.doi.org/10.7717/peerj.12573>. 
  37. Tulewicz-Marti E, Szwarc P, Więcek M et al. Effect of Intravenous Iron Administration on Bone Mineral and Iron Homeostasis in Patients with Inflammatory Bowel Disease-Results of a Prospective Single-Centre Study. J Pers Med 2023; 13(3): 458. Available on DOI: <http://dx.doi.org/10.3390/jpm13030458>. 
  38. Rioux FM, LeBlanc CP. Iron supplementation during pregnancy: what are the risks and benefits of current practices? Appl Physiol Nutr Metab 2007; 32(2): 282–288. Available on DOI: <http://dx.doi.org/10.1139/H07–012>. 
  39. Isidori A, Borin L, Elli E et al. Iron toxicity – Its effect on the bone marrow. Blood Rev 2018; 32(6): 473–479. Available on DOI: <http://dx.doi.org/10.1016/j.blre.2018.04.004>. 
  40. Abbaspour N, Hurrell R, Kelishadi R. Review on iron and its importance for human health. J Res Med Sci 2014; 19(2): 164–174.
  41. Frewin R, Hensen A, Provan D. ABC of clinical haematology. Iron deficiency anaemia. BMJ 1997; 314(7077): 360–363. Available on DOI: <http://dx.doi.org/10.1136/bmj.314.7077.360>. 
  42. Jeney V. Clinical Impact and Cellular Mechanisms of Iron Overload-Associated Bone Loss. Front Pharmacol 2017; 8: 77. Available on DOI: <http://dx.doi.org/10.3389/fphar.2017.00077>. 
  43. Sun L, Guo W, Yin C, et al. Hepcidin deficiency undermines bone load-bearing capacity through inducing iron overload. Gene 2014; 543(1): 161–165. Available on DOI: <http://dx.doi.org/10.1016/j.gene.2014.02.023>.
  44. Liu LL, Liu ZR, Cao LJ et al. Iron accumulation induced by hepcidin1 knockout accelerates the progression of aging osteoporosis. J Orthop Surg Res 2024; 12;19(1): 59. Available o DOI: <http://doi: 10.1186/s13018-024-04535-z>.
Štítky
Biochémia Detská gynekológia Detská rádiológia Detská reumatológia Endokrinológia Gynekológia a pôrodníctvo Interné lekárstvo Ortopédia Praktické lekárstvo pre dospelých Rádiodiagnostika Rehabilitácia Reumatológia Traumatológia Osteologie
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#