en POLSKI
eISSN: 2300-8660
ISSN: 0031-3939
Pediatria Polska - Polish Journal of Paediatrics
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2024
vol. 99
 
Share:
Share:
Review paper

Revisiting copper and zinc in end-stage renal disease patients

Maha Abd Sahab
1
,
Basim Abd Al-Raheem
1
,
Khalid Farouk
2
,
Hussein Kadhem Al-Hakeim
1
,
Maha Abd Al-Sahab
3

  1. Department of Chemistry, Faculty of Science, University of Kufa, Iraq
  2. Department of Chemistry, Faculty of Science, University of Anbar, Iraq
  3. College of Science, Kufa University, Najaf, Iraq
Pediatr Pol 2024; 99 (2): 143-148
Online publish date: 2024/06/27
Article file
- Revisiting copper.pdf  [0.28 MB]
Get citation
 
PlumX metrics:
 

END-STAGE RENAL DISEASE

The kidneys play a vital role in the elimination of blood wastes and excess fluid via the process of urine excretion. The body may experience the accumulation of fluid, electrolytes, and waste substances at toxic levels when the kidneys fail to filter these components [1]. When the gradual decline in renal function reaches an advanced stage, it reaches end-stage renal disease (ESRD), which is also known as end-stage kidney disease or kidney failure [1, 2]. National Kidney Foundation has categorised kidney disease into 5 distinct stages. Stage 5 specifically pertains to those with a glomerular filtration rate (GFR) of less than 15 ml per minute per 1.73 m2 of body surface area or those who need dialysis irrespective of their GFR, including refractory oedema, uraemia, acidosis, hyperkalaemia, and/or unacceptable symptoms [3, 4]. Decreased or missing kidney function leads to a range of maladaptive alterations, such as extracellular volume excess, resulting in fluid retention, anaemia, disturbances in bone and mineral metabolism, dyslipidaemia, and protein energy deficit [5].
Patients with ESRD require either dialysis or a kidney transplant in order to survive. In order to effectively address symptoms and optimise the overall quality of life throughout the remaining period, it is essential to include conservative treatment measures [6]. Multiple studies have shown a reduction in zinc levels in the blood plasma of patients with chronic renal insufficiency who are undergoing conservative treatment or haemodialysis (HD) [7, 8]. Some authors have reported that zinc levels in erythrocytes are within the normal range in comparable patient groups, whereas others report higher levels [9, 10]. The primary contributors to the trace element decrease in HD patients are urine losses and losses to the dialysate [11, 12].
The influence of renal sickness on trace element concentration in uraemic patients is mostly determined by the severity of the condition [13]. Individuals diagnosed with ESRD often experience fluid retention, causing hypertension, heart ventricular failure, and subsequent cardiovascular incidents [14]. This fluid retention may cause the abnormal distribution of the metal ions between plasma and tissue reservoirs.

TRACE ELEMENTS

Trace elements are a set of metals that naturally occur in biological systems at low concentrations, such as zinc (Zn), iron (Fe), and copper (Cu) [15]. As a result of urbanisation and industrialisation, the amount of trace elements that individuals contribute to the environment is increasing overall [16], and because these elements do not biodegrade, they move through the environment in biogeochemical cycles. The concern around trace elements arises from their significant capacity to inflict damage upon organisms, even in small concentrations. Trace elements have a multitude of important functions inside the human body, including activating enzymatic reactions by enhancing the conversion of substrate molecules into certain end products [17]. The idea that the trace element status affects the probability of undesirable clinical outcomes seems worthy of inquiry because both trace element deficiencies and excesses may be treatable. To clarify the clinical significance and long-term implications of these trace element abnormalities, more research is required [18]. Among the important trace elements, we focused in the present review on the levels of serum copper and zinc in ESRD patients who underwent haemodialysis.

1-ZINC

Zinc is a vital trace metal that serves as a cofactor for more than 300 distinct enzymes involved in cell metabolism, growth, and development [19]. Zinc is involved in the metabolic processes of proteins, carbohydrates, lipids, and energy via its participation as a constituent of many enzymes [19]. It is essential for the synthesis of proteins and DNA, as well as for insulin action and the proper functioning of liver metabolism [20].
Patients undergoing HD appear to have lower zinc levels than the general population [21, 22]. Zinc deficiency is prevalent in undeveloped nations [23], and it is linked to immunological deficiencies that have defective T-cell function, poor phagocytosis, inappropriate cytokine production, and delayed wound healing [24], which causes an elevated risk of infection seen in HD patients. Anorexia in HD patients may also be brought on by or contributed to by zinc deficiency [25]. Anorexia and a diminished sense of taste and smell are just a few of the uremic symptoms that are allegedly associated with zinc deficiency in HD patients. Zinc shortage in individuals with uraemia may arise because of reduced food intake and intestinal absorption, with increased losses of zinc from the body [26]. Researchers believe that the hormone leptin might be used as a tool to investigate the physiological pathways that are responsible for zinc deficiency- induced anorexia [27]. The association between zinc and leptin has been widely found in HD patients [28]. The possible mechanisms of the alteration of zinc in kidney diseases are presented in Figure 1.
Many causes of zinc deficiency are presented in Figure 1, including diet restriction, albuminuria, loss by HD, malabsorption, and drug interactions [22, 29, 30]. To buffer the zinc deficiency, the zinc will be released from bone and tissues to supply the need for zinc to the erythrocyte formation. The role of dialysis treatment on zinc metabolism needs to be considered because of the presence of an inverse relationship between erythrocytes and zinc [31]. This negative association, as a measure of dialysis adequacy, could indicate a redistribution of zinc due to the loss of this element during the dialysis treatment [30]. However, studies have not shown a loss of zinc during haemodialysis, and a rise in plasma zinc was discovered during the dialysis procedure, which could be because of haem concentration [32], besides the leaked zinc from the haemolysed erythrocytes [33].
Zinc is the cofactor of many enzymes, including superoxide dismutase, which catalyses the elimination of intracellular free radicals, and metallothionein, which traps free radicals and prevents lipid peroxidation [34]. Numerous investigations have revealed that CKD patients who were receiving either conservative treatment or dialysis had a significant frequency of zinc deficiency [35]. In several illness situations, such as ESRD patients undergoing HD, the risk of zinc deficiency is extremely high [26]. The causes of zinc shortage that have been linked to previous studies include protein-energy malnutrition, inadequate dietary intake, abnormalities of absorption and transport, and increased excretion [29]. The plasma zinc concentration noticeably drops in ESRD patients who are receiving HD therapy. This decrease often leads to a genuine zinc deficiency, regardless of the specific type of HD being administered [36].
In one study, over half of the HD group had a major zinc deficit, and zinc levels were significantly lower in the HD group than in the control group [37]. The precise mechanism underlying the reduction of zinc concentration in patients with CKD remains uncertain. Patients on HD did not have low reported whole-body zinc levels, in contrast to low serum zinc levels [38]. Several factors have observed decreases in nutritional intake seem to be associated with a limitation on protein, while inadequate gastrointestinal absorption may be caused by a deficiency in vitamin D [39] or interactions between drugs, such as the use of phosphate binders [40]. An additional potential factor contributing to diminished zinc levels in patients with CKD is excessive urinary excretion of zinc. Patients with CKD had lower levels of zinc in their bloodstream due to the higher levels of excretion in their zinc compared to the control group [41]. Importantly, zinc and copper levels were inversely correlated, suggesting that taking zinc acetate orally may increase the risk of copper insufficiency. After the prescription of zinc acetate, it may be prudent to check both zinc and copper levels every month [42].
Zinc is transported by albumin [43]. Hypoalbuminaemia or a systemic inflammatory response both caused low plasma zinc concentration, suggesting that zinc insufficiency may have been misdiagnosed by interpreting low plasma zinc concentration as being caused by hypoalbuminaemia [44].

2-COPPER

In the intestines, copper is absorbed and then transferred to the liver bound with albumin. Copper is transported to various tissues in a second phase after being processed in the liver. The majority of the copper in the blood is carried by the protein ceruloplasmin, which is involved in copper transport in the liver [45]. Wheat, barley, fish, sunflower, and chicken are the finest dietary sources of copper for the human body [46]. Copper is an element that is essential to the functioning of several enzymes, including cytochrome oxidase, monoamine oxidase, and superoxide dismutase (SOD) [47].
Furthermore, because of its presence in many enzymes, copper is engaged in a wide range of metabolic reactions. The inclusion of copper in SOD, for example, aids in the conversion of superoxide to hydrogen peroxide and oxygen [48]. Copper is required for the haematopoietic and nervous systems as well as musculoskeletal. The gene expression during neural development and apoptosis, because it shows the molecular functions of copper and zinc [49], demonstrates that copper affects the protein structure of neurons to promote neurotransmission. Copper could alter the neuroproteostasis of CNS neurons, changing the excitability of those neurons [50]. It is required for bone growth and synthesis, aids in the incorporation of Fe into haemoglobin, aids in the absorption of Fe from the gastrointestinal tract, and facilitates the transport of Fe from tissues to plasma [46]. Copper deficiency is seldom seen in individuals who are in good health; however, it may manifest in children. The copper deficiency arises mostly as a result of impaired copper absorption [46]. A copper-rich diet can lead to the accumulation of this microelement in the kidney causing proximal tube necrosis caused by oxidative stress [51] and damage to cells, leading to a reduction in kidney function [52]. Figure 2 illustrates the harmful effects of hypercupraemia on the kidney in addition to the mechanisms of loss of Fe in HD patients.
The expression of copper transporter 1 (CTR1) in renal tissues was elevated in patients with renal fibrosis, causing anaemia. Transforming growth factor β (TGF-β)- treated renal tubular epithelial cells and fibroblasts demonstrated comparable outcomes [53, 54]. Mechanistically, TGF-β signalling upregulated CTR1 and induced increased intracellular copper influx. The activation of lysyl oxidase was seen as a result of elevated levels of intracellular copper ions, leading to an increase in the cross-linking of collagen and elastin [53, 54]. Furthermore, this phenomenon was shown to contribute to the development of renal fibrosis. The results of the alterations in serum copper and zinc levels are subject to conflicting interpretations. Some researchers found decreased blood levels of zinc and copper in ESRD patients [55]. Previous studies on uremic patients have shown inconsistent results about low levels of serum zinc and copper [56]. The first measure to decrease copper in ESRD patients is through treatment of inflammation to reduce the ceruloplasmin that acts as an acute phase reactant protein [57]. Conversely, people with ESRD have low zinc, but high serum copper [58, 59]. In patients on HD with low serum zinc concentrations, leukopaenia, which can be megaloblastic or sideroblastic, and copper deficiency may coexist [60]. However, compared to healthy controls, HD patients’ average blood concentrations of physiologically significant trace elements like copper and zinc were significantly different [61]. Besides organelle malfunction and lipid peroxidation, the disruption of copper homeostasis results in the toxic alkenal formation, including the production of 4-hydroxynonenal that acts as an inhibitor of pyruvate dehydrogenase and α-keto-glutarate dehydrogenase (α-KGDH) [62, 63]. When the α-KGDH complex is inhibited, cytochrome c is released from mitochondria, caspase-3 is activated, and necrotic cells die [64]. The recommended daily allowance for copper is 900 µg per day. To prevent copper deficiency or toxicity, individuals need to consume an appropriate quantity of copper in their diet [65]. To maintain health, the body needs modest amounts of copper from meals to prevent anaemia associated with renal diseases [66]. However, in children treated with HD, copper supplements are used to adjust the haematological abnormalities [42]. Normally, the liver excretes excess copper by releasing it in bile, but a buildup of copper is dangerous and, if left untreated, can cause brain damage, liver failure, or even death [67].

CONCLUSIONS

In ESRD patients, abnormal zinc and copper homeostasis are widely recorded, which may lead to an increase in the probability of a poor prognosis due to the importance of these trace elements in many biological processes. Therefore, it is important to take therapeutic measures, such as screening and treatment, to mitigate the adverse effects caused by abnormal levels.

Disclosures

  • Institutional review board statement: Not applicable.
  • Assistance with the article: None.
  • Financial support and sponsorship: None.
  • Conflicts of interest: None.
    REFERENCES
  • 1. Twaij BAAR, Al-Hakeim HK, Altufaili MF. Review on the neuron damage parameters in patients with end-stage renal disease. J Popul Ther Clin Pharmacol 2023; 30: 408-420.
    2. Chertow GM, Vart P, Jongs N, et al. Effects of dapagliflozin in stage 4 chronic kidney disease. J Am Soc Nephrol 2021; 32: 2352.
    3. Roberts PR, Green D. Arrhythmias in chronic kidney disease. Heart 2011; 97: 766-773.
    4. Moss AH. Ethical considerations in the care of dialysis patients: the new paradigm. Handbook of dialysis therapy, Elsevier 2023, 606-612.
    5. Romagnani P, Remuzzi G, Glassock R, et al. Chronic kidney disease. Nat Rev Dis Prim 2017; 3: 1-24.
    6. Jakimowicz T, Przywara S, Turek J, et al. Five year outcomes in patients with end stage renal disease who received a bioengineered human acellular vessel for dialysis access. EJVES Vasc Forum 2022; 54: 58-63.
    7. Fukushima T, Horike H, Fujiki S, Kitada S, Sasaki T, Kashihara N. Zinc deficiency anemia and effects of zinc therapy in maintenance hemodialysis patients. Ther Apher Dial 2009; 13: 213-219.
    8. Guo CH, Wang CL, Chen PC, Yang TC. Linkage of some trace elements, peripheral blood lymphocytes, inflammation, and oxidative stress in patients undergoing either hemodialysis or peritoneal dialysis. Perit Dial Inter 2011; 31: 583-591.
    9. Montesa MP, Rico MG, Salguero MS, et al. Study of oxidative stress in advanced kidney disease. Nefrologia 2009; 29: 464-473.
    10. Noleto Magalhães RC, Guedes Borges de Araujo C, Batista de Sousa Lima V, Machado Moita Neto J, do Nascimento Nogueira N, do Nascimento Marreiro D. Nutritional status of zinc and activity superoxide dismutase in chronic renal patients undergoing hemodialysis. Nutr Hosp 2011; 26: 1456-1461.
    11. Wakino S. Trace elements and their management in dialysis patients – pathophysiology and clinical manifestations. Kidney Dial 2023; 3: 274-296.
    12. Prodanchuk M, Makarov O, Pisarev E, Sheiman B, Kulyzkiy M. Disturbances of trace element metabolism in ESRD patients receiving hemodialysis and hemodiafiltration. Cent European J Urol 2014; 66: 472-476.
    13. Almeida A, Gajewska K, Duro M, Costa F, Pinto E. Trace element imbalances in patients undergoing chronic hemodialysis therapy – report of an observational study in a cohort of Portuguese patients. J Trace Elem Med Biol 2020; 62: 126580.
    14. Law JP, Pickup L, Pavlovic D, Townend JN, Ferro CJ. Hypertension and cardiomyopathy associated with chronic kidney disease: epidemiology, pathogenesis and treatment considerations. J Hum Hypertens 2023; 37: 1-19.
    15. Ali W, Mao K, Zhang H, et al. Comprehensive review of the basic chemical behaviours, sources, processes, and endpoints of trace element contamination in paddy soil-rice systems in rice-growing countries. J Hazard Mater 2020; 397: 122720.
    16. Mahapatra SR, Nimmy P (ed.). Groundwater contamination with heavy metals in Chennai city, India – a threat to the human population. IOP Conference Series: Earth and Environmental Science 2021, IOP Publishing.
    17. Twaij BAAR, Mashkour MS, Al-Hakeim HK. Study and determination of aryl hydrocarbon receptor with some trace elements in fuel filling station workers. J Petrol Res Stud 2020; 10: 233-251.
    18. Zhu X, Zhang Y, Han J, Xiao D, Zhu D, Ou Y. Effects of chronic kidney disease on levels of oxidative stress and trace elements. System Rev Pharma 2022; 13: 128-134.
    19. Islam MR, Akash S, Jony MH, et al. Exploring the potential function of trace elements in human health: a therapeutic perspective. Mol Cell Biochem 2023; 478: 2141-2171.
    20. Chasapis CT, Ntoupa PSA, Spiliopoulou CA, Stefanidou ME. Recent aspects of the effects of zinc on human health. Arch Toxicol 2020; 94: 1443-1460.
    21. Neto L, Bacci M, Sverzutt L, Costa M, Alves B, Fonseca F. The role of zinc in chronic kidney disease patients on hemodialysis: a systematic review. Health 2016; 8: 344.
    22. Esfahani ST, Hamidian MR, Madani A, et al. Serum zinc and copper levels in children with chronic renal failure. Pediatr Nephrol 2006; 21: 1153-1156.
    23. Jeng SS, Chen YH. Association of zinc with anemia. Nutrients 2022; 14: 4918.
    24. Akar-Ghibril N. Defects of the innate immune system and related immune deficiencies. Clin Rev Allergy Immunol 2022; 63: 36-54.
    25. Humudat YR, Al-Naseri SK. Heavy metals in dialysis fluid and blood samples from hemodialysis patients in dialysis centers in Baghdad, Iraq. J Health Pollut 2020; 10: 200901.
    26. Mafra D, Cuppari L, Cozzolino SM. Iron and zinc status of patients with chronic renal failure who are not on dialysis. J Ren Nutr 2002; 12: 38-41.
    27. Ott ES, Shay NF. Zinc deficiency reduces leptin gene expression and leptin secretion in rat adipocytes. Exp Biol Med (Maywood) 2001; 226: 841-846.
    28. Aranha LN, Lobo JC, Stockler-Pinto MB, de Oliveira Leal V, Torres JPM, Mafra D. Relationship between zinc levels and plasma leptin in hemodialysis patients. J Trace Elem Med Biol 2012; 26: 238-242.
    29. Bozalioğlu S, Özkan Y, Turan M, Şimşek B. Prevalence of zinc deficiency and immune response in short-term hemodialysis. J Trace Elem Med Biol 2005; 18: 243-249.
    30. Davenport A. Prevalence and determinants of low plasma zinc levels in adult peritoneal dialysis patients. J Trace Elem Med Biol 2023; 78: 127171.
    31. Rodic S, McCudden C, van Walraven C. Relationship between plasma zinc and red blood cell zinc levels in hospitalized patients. J Appl Lab Med 2022; 7: 1412-1423.
    32. Palmer BF. Evaluation and treatment of sexual dysfunction. Handbook of Dialysis Therapy, Elsevier 2023, 507-512.
    33. Sam R, Haghighat L, Kjellstrand CM, Ing TS. Hemolysis during hemodialysis. Handbook of Dialysis Therapy, Elsevier 2008, 457.
    34. Chasapis CT, Ntoupa PSA, Spiliopoulou CA, Stefanidou ME. Recent aspects of the effects of zinc on human health. Arch Toxicol 2020; 94: 1443-1460.
    35. Kiziltas H, Ekin S, Erkoc R. Trace element status of chronic renal patients undergoing hemodialysis. Biol Trace Elem Res 2008; 124: 103-109.
    36. Hasanato RM. Assessment of trace elements in sera of patients undergoing renal dialysis. Saudi Med J 2014; 35: 365-370.
    37. Dashti-Khavidaki S, Khalili H, Vahedi SM, Lessan-Pezeshki M. Serum zinc concentrations in patients on maintenance hemodialysis and its relationship with anemia, parathyroid hormone concentrations and pruritus severity. Saudi J Kidney Dis Transpl 2010; 21: 641-645.
    38. Abdulla M. How adequate is plasma zinc as an indicator of zinc status? Prog Clin Biol Res 1983; 129: 171-183.
    39. Kambe T, Tsuji T, Hashimoto A, Itsumura N. The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 2015; 95: 749-784.
    40. Takagi K, Masuda K, Yamazaki M, et al. Metal ion and vitamin adsorption profiles of phosphate binder ion-exchange resins. Clin Nephrol 2010; 73: 30-35.
    41. Damianaki K, Lourenco JM, Braconnier P, et al. Renal handling of zinc in chronic kidney disease patients and the role of circulating zinc levels in renal function decline. Nephrol Dial Transplant 2020; 35: 1163-1170.
    42. Nishime K, Kondo M, Saito K, Miyawaki H, Nakagawa T. Zinc burden evokes copper deficiency in the hypoalbuminemic hemodialysis patients. Nutrients 2020; 12: 577.
    43. Lu J, Stewart AJ, Sadler PJ, Pinheiro TJ, Blindauer CA. Albumin as a zinc carrier: properties of its high-affinity zinc-binding site. Biochem Soc Trans 2008; 36: 1317-1321.
    44. Duncan A, Yacoubian C, Watson N, Morrison I. The risk of copper deficiency in patients prescribed zinc supplements. J Clin Pathol 2015; 68: 723-725.
    45. Clarkson A, Paine S, Martín-Tereso J, Kendall N. Copper physiology in ruminants: trafficking of systemic copper, adaptations to variation in nutritional supply and thiomolybdate challenge. Nutr Res Rev 2020; 33: 43-49.
    46. Al-Fartusie FS, Mohssan SN. Essential trace elements and their vital roles in human body. Indian J Adv Chem Sci 2017; 5: 127-136.
    47. Angelova M, Asenova S, Nedkova V, Koleva-Kolarova. Copper in the human organism. Trakia J Sci 2011; 9: 88-98.
    48. Ali SS, Ahsan H, Zia MK, Siddiqui T, Khan FH. Understanding oxidants and antioxidants: classical team with new players. J Food Biochem 2020; 44: e13145.
    49. Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernandez-Rodriguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. BioMetals 2022; 35: 395-427.
    50. Washington-Hughes CL, Roy S, Seneviratne HK, et al. Atp7b- dependent choroid plexus dysfunction Causes transient copper deficit and metabolic changes in the developing mouse brain. PLoS Gen 2023; 19: e1010558.
    51. Abolaji AO, Fasae KD, Iwezor CE, Aschner M, Farombi EO. Curcumin attenuates copper-induced oxidative stress and neurotoxicity in Drosophila melanogaster. Toxicol Rep 2020; 7: 261-268.
    52. Ay A, Alkanli N, Ustundag S. Investigation of the relationship between IL-18 (- 607 C/A), IL-18 (- 137 G/C), and MMP-2 (- 1306 C/T) gene variations and serum copper and zinc levels in patients diagnosed with chronic renal failure. Biol Trace Elem Res 2022; 200: 2040-2052.
    53. Niu Y, Zhang Y, Zhu Z, et al. Elevated intracellular copper contributes a unique role to kidney fibrosis by lysyl oxidase mediated matrix crosslinking. Cell Death Dis 2020; 11: 211.
    54. Cheng F, Peng G, Lu Y, et al. Relationship between copper and immunity: the potential role of copper in tumor immunity. Front Oncol 2022; 12: 1019153.
    55. Anees M, Mumtaz A, Frooqi S, Ibrahim M, Hameed F. Serum trace elements (aluminium, copper, zinc) in hemodialysis patients. Biomedica 2011; 27: 106-110.
    56. Liu Y, Tang R, Xu Q, et al. High blood Cu/Zn ratio is associated with nutritional risk in patients undergoing maintenance hemodialysis. Biol Trace Elem Res 2022; 200: 4977-4987.
    57. Nakamura H, Ueda M, Anayama M, Makino Y, Nagasawa M. Short-term effects of roxadustat on serum copper and iron changes in a peritoneal dialysis patient. CEN Case Rep 2023; 12: 292-296.
    58. Stojsavljević A, Ristić-Medić D, Krstić D, et al. Circulatory imbalance of essential and toxic trace elements in pre-dialysis and hemodialysis patients. Biol Trace Elem Res 2021; 200: 3117-3312.
    59. Cedeño Y, Miranda M, Orjales I, et al. Serum concentrations of essential trace and toxic elements in healthy and disease-affected dogs. Animals 2020; 10: 1052.
    60. Neuberger J. Liver tests. In: Hirschfieldm GM, Gill P, Neuberger J (eds.). The Liver in systemic disease: a clinician’s guide to abnormal liver tests. John Wiley & Sons Ltd., NY 2023, 1-18.
    61. Stojsavljević A, Ristić-Medić D, Krstić D, et al. Circulatory imbalance of essential and toxic trace elements in pre-dialysis and hemodialysis patients. Biol Trace Elem Res 2022; 200: 3117-3125.
    62. Zischka H, Borchard S. Mitochondrial copper toxicity with a focus on Wilson disease. In: Kerkar N, Roberts EA. Clinical and Translational Perspectives on WILSON DISEASE. Elsevier 2019, 65-75.
    63. Sheline CT, Choi DW. Cu2+ toxicity inhibition of mitochondrial dehydrogenases in vitro and in vivo. Ann Neurol 2004; 55: 645-653.
    64. Huang HM, Ou HC, Xu H, Chen HL, Fowler C, Gibson GE. Inhibition of alpha-ketoglutarate dehydrogenase complex promotes cytochrome c release from mitochondria, caspase-3 activation, and necrotic cell death. J Neurosci Res 2003; 74: 309-317.
    65. Eljazzar S, Abu-Hijleh H, Alkhatib D, et al. The role of copper intake in the development and management of type 2 diabetes: a systematic review. Nutrients 2023; 15: 1655.
    66. Nazari-Taloki Z, Salehifar E, Makhlough A, Dashti-Khavidaki S. Effect of recommended dietary intake versus higher doses of supplemental zinc on iron and copper deficiency anemia among patients with chronic kidney diseases, a double-blinded, randomized clinical trial. Pharm Sci 2023; 29: 495-503.
    67. Liu T, Liu Y, Zhang F, Gao Y. Copper homeostasis dysregulation promoting cell damage and the association with liver diseases. Chin Med J 2023; 136: 1653-1662.
    Copyright: © 2024 Polish Society of Paediatrics. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.