Next Article in Journal
Role of Dietary Fiber and Energy Intake on Gut Microbiome in Vegans, Vegetarians, and Flexitarians in Comparison to Omnivores—Insights from the Nutritional Evaluation (NuEva) Study
Next Article in Special Issue
Effect of Menaquinone-7 Supplementation on Arterial Stiffness in Chronic Hemodialysis Patients: A Multicenter Randomized Controlled Trial
Previous Article in Journal
Antioxidant Compounds from Edible Mushrooms as Potential Candidates for Treating Age-Related Neurodegenerative Diseases
Previous Article in Special Issue
Chronic Kidney Disease—State of Either “Too Much” or “Too Little”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Further Evidence on Trace Element Imbalances in Haemodialysis Patients—Paired Analysis of Blood and Serum Samples

1
LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
2
Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padova, Italy
3
FP-ENAS—Fernando Pessoa Energy, Environment and Health Research Unit, Fernando Pessoa University, 4249-004 Porto, Portugal
4
Laboratório de Análises Clínicas Dra. Matilde Sampaio, 5200-216 Mogadouro, Portugal
5
Laboratório de Análises Clínicas Vale do Sousa, 4560-547 Penafiel, Portugal
6
Department of Environmental Health, ESS, Polytechnic of Porto, 4200-072 Porto, Portugal
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(8), 1912; https://doi.org/10.3390/nu15081912
Submission received: 10 February 2023 / Revised: 5 April 2023 / Accepted: 14 April 2023 / Published: 15 April 2023
(This article belongs to the Special Issue Micronutrient Deficiencies and Chronic Kidney Disease)

Abstract

:
Previous studies have shown that haemodialysis patients have an increased risk of trace element imbalances. Most studies have determined the concentration of trace elements in serum only, but most trace elements are not uniformly distributed between plasma and blood cells, which justifies separate analysis of the different compartments. In this study, we determined both the serum and whole blood concentration of a wide panel of trace elements (Li, B, Mn, Co, Ni, Cu, Zn, Se, Rb, Sr, Mo, Cd, Pb) in haemodialysis patients and compared them with those of a control group. Whole blood and serum samples were collected during routine laboratory testing of patients undergoing chronic haemodialysis. For comparison purposes, samples from individuals with normal renal function were also analysed. Statistically significant differences (p < 0.05) were found between the two groups for whole blood concentrations of all analysed elements except Zn (p = 0.347). For serum, the difference between groups was statistically significant for all elements (p < 0.05). This study confirms that patients on haemodialysis tend to present significant trace element imbalances. By determining the concentration of trace elements in both whole blood and serum, it was shown that chronic haemodialysis may affect intra- and extracellular blood compartments differently.

1. Introduction

Chronic kidney disease (CKD) is defined as abnormalities of kidney structure or function present for more than 3 months, with implications for the individual’s health [1]. CKD affects more than 10% of the general population worldwide, which corresponds to more than 800 million individuals [2]. It is more prevalent in older, female individuals and is usually accompanied by several comorbidities, such as diabetes, hypertension and cardiovascular diseases [2,3].
The majority of CKD cases are irreversible and have a lifelong progression. Thus, treatment is directed at slowing the progression to kidney failure [1]. Kidney failure is characterised by a glomerular filtration rate (GFR) < 15 mL/min/1.73 m2 and requires renal replacement therapy (RRT), most often haemodialysis (HD), peritoneal dialysis or kidney transplantation [1]. In 2010, 2.6 million individuals received RRT worldwide, and it is estimated that at least 4.9 million individuals will need RRT by 2030 [4].
Haemodialysis is the most common modality of RRT [5,6]. However, while HD offers years of life, it is a chronic therapy that affects the lifestyle, eating habits and overall quality of life of patients [7,8,9] and their families and caregivers [10,11,12].
In HD, a machine pumps blood through a tube system from the patient’s vein to the dialyser (“artificial kidney”), where the blood is cleared through an artificial semipermeable membrane that separates it from the dialysate, then returns it to the patient’s body [13,14]. The semipermeable membrane is the key component of the entire system, regardless of which of the two the basic processes of solute and fluid movement are used: (i) diffusive clearance, where solutes move down their concentration gradient from areas of high concentration to low concentration, and (ii) convective clearance (hemofiltration or ultrafiltration), where a higher hydrostatic pressure in the blood compartment than in the dialysate compartment is used to promote the passage of blood fluid into the dialysate, with the solutes moving along with the water, a modality that requires the concomitant administration of a replacement fluid to compensate for the large fluid removal during hemofiltration [13,14].
Previous studies have shown that HD patients are at an increased risk of trace element imbalances [15,16,17,18,19,20,21,22,23]. While the concentration of essential trace elements may be decreased due to the inflammatory nature of the disease [24,25,26], lower food intake due to uremic anorexia or dietary restrictions [27,28,29] or losses during dialysis sessions [30], the concentration of non-essential/toxic trace elements may be increased due to loss of ability to excrete them in urine [31], among other factors.
Most studies on trace element imbalances in HD patients have analysed their serum/plasma concentration [15,16,17,18,19,20], with only a few focusing on whole blood analysis [22,23]. However, in most cases, circulating trace elements are not uniformly distributed between plasma and cellular blood components. For example, only about 4% of circulating Mn is present in plasma, with 66% found in erythrocytes and the remaining 30% in leucocytes and platelets [32]. Another example is Pb, where approximately 99% of circulating Pb is found in erythrocytes [33]. This means that changes observed in the serum of HD patients may not be present in whole blood or its cellular components, and vice versa.
Previously, we observed lower serum concentrations of Mn, Cu, Zn, Se, Rb and Ba and higher concentrations of Al, Co, Ni, Sr, Mo, Cd and Pb in a group of HD patients compared to a control group (individuals without renal impairment) [34]. The present study aimed to further investigate this issue by measuring both serum and whole blood concentrations of several essential and non-essential/toxic trace elements in a cohort of HD patients and comparing them to a control group.

2. Materials and Methods

2.1. Study Design and Sample Collection

This work is a comparative study of trace element concentrations in serum and whole blood of HD patients and a control group of individuals without renal impairment. Whole blood (n = 108) and serum (n = 88) samples were collected during routine laboratory testing from individuals undergoing regular HD therapy at a Dialysis Centre in northern Portugal. The control group was established by using whole blood (n = 59) and serum (n = 20) samples from apparently healthy individuals who attended the same Clinical Laboratory and met the standard laboratory analytical criteria for normal kidney function.
Whole blood samples were collected by conventional venepuncture into BD (Franklin Lakes, NJ, USA) Vacutainer™ polyethylene terephthalate (PET) tubes (K2EDTA as anticoagulant) or Greiner Bio-One (Madrid, Spain) VACUETTE® PET tubes (sodium heparin as anticoagulant), special for trace element analysis, and kept refrigerated until analysis. Serum samples were obtained by collecting blood through conventional venepuncture into VACUETTE® Z Trace Elements no-additive PET tubes. After allowing the specimen to clot for 30 min, the serum was separated into previously decontaminated Eppendorf tubes and kept refrigerated until analysis.
Data anonymisation was ensured by removing all patient information except sex and age. The study was approved by the Ethics Committee of the Faculty of Pharmacy of the University of Porto (Parecer Nº 38-06-2019).

2.2. Reagents

All solutions were prepared with ultrapure water (>18.2 MΩ·cm at 25 °C) obtained with a Sartorius Arium® pro water purification system (Gottingen, Germany). Nitric acid (HNO3, 67–69% w/w TraceMetal® Grade) was obtained from Fisher Scientific (Leicestershire, UK). Triton X-100, butanol (>99.0%) and multi-element stock solutions (10 mg/L) Periodic Table Mix 1, 2 and 3 were obtained from Sigma-Aldrich (St. Louis, MO, USA). Copper and Zn single-element stock solutions (1000 mg/L) were obtained from SCP Science (Quebec, QC, Canada). All laboratory ware (bottles, tubes, volumetric flasks) was made of polypropylene or high-density polyethylene (HDPE) and was properly decontaminated by immersion in a 10% v/v HNO3 solution for at least 24 h, followed by abundant rinsing with ultrapure water and drying at room temperature under dust-free conditions.

2.3. Laboratory Procedures

Whole blood and serum samples were analysed at the Laboratory of Applied Chemistry (Trace Element Analysis Unit) of the Faculty of Pharmacy, University of Porto, Portugal, using properly validated inductively coupled plasma mass spectrometry (ICP-MS) analytical procedures. The instrument was an iCAP™ Q (Thermo Fisher Scientific, Bremen, Germany), equipped with a Meinhard® (Golden, CO) TQ+ quartz concentric nebuliser, a Peltier cooled, high purity quartz, baffled cyclonic spray chamber and a demountable quartz torch with a 2.5 mm i.d. quartz injector. The interface consisted of two Ni cones (sampler and skimmer). High-purity argon (99.9997%) supplied by Gasin (Matosinhos, Portugal) was used both as nebuliser gas and plasma gas. Prior to each analytical run, the instrument was tuned for maximum sensitivity and signal stability and for minimal formation of oxides and double charged ions. The main operating parameters of the ICP-MS instrument were nebuliser gas flow, 1.16 L/min; auxiliary gas flow, 0.79 L/min; plasma gas flow, 13.9 L/min; radio frequency generator power, 1550 W; dwell time, 10 ms.
For serum analysis, a procedure based on Goullé et al. [35] was used. Briefly, samples were diluted 1:10 with a diluent solution containing 0.65% v/v HNO3, 0.01% v/v Triton X-100, 0.5% v/v butanol and internal standards (IS) at 10 µg/L (added by proper dilution of Periodic Table Mix 3 for ICP). For whole blood analysis, a similar procedure based on Goullé et al. [35] was used. Briefly, samples were diluted 1:15 with a diluent solution containing 0.65% v/v HNO3, 0.1% v/v Triton X-100, 0.5% v/v butanol and IS at 10 µg/L (added by proper dilution of Periodic Table Mix 3 for ICP).
For Li, B, Mn, Co, Ni, Se, Rb, Sr, Cd and Pb, an 8-point calibration curve (1, 5, 10, 25, 50, 100, 250 and 500 µg/L) was generated with standard solutions prepared by appropriate dilution of Periodic Table Mix 1 in 2% HNO3. For Mo, an 8-point calibration curve (0.05, 0.25, 0.5, 1.25, 2.5, 5, 12.5 and 25 µg/L) was generated with standard solutions prepared by appropriate dilution of Periodic Table Mix 2 in 2% HNO3. For Cu and Zn, a 5-point calibration curve (0.5, 1.0, 1.5, 2.0 and 5.0 mg/L) was generated with mixed standard solutions prepared by appropriate dilution of single-element standard stock solutions in 2% HNO3. Calibration solutions were then diluted 1:10 (for serum analysis) or 1:15 (for whole blood analysis) with the diluent solution, as the samples. The elemental isotopes 7Li, 11B, 55Mn, 59Co, 60Ni, 65Cu, 66Zn, 82Se, 85Rb, 88Sr, 98Mo, 111Cd, 206Pb, 207Pb and 208Pb were measured for analytical determinations and 89Y, 141Pr and 159Tb were monitored as IS.
After complete mixing on a vortex mixer, the diluted samples and calibration standards were presented to the ICP-MS instrument using a CETAC ASX-520 auto sampler (Teledyne CETAC Technologies, Omaha, NE). Samples were analysed in random order to avoid sequence effects. For analytical quality assurance, repeated analysis (at the beginning, middle and end of each analytical run) of Seronorm™ Trace Elements Serum L-1 and L-2 and Seronorm™ Trace Elements Whole Blood L-1, L-2 and L-3 (obtained from SERO AS, Billingstad, Norway) was performed. The results obtained are presented in Supplementary Materials (Table S1).

2.4. Statistical Analysis

Statistical analysis was performed using SPSS Statistics v.27.0 (IBM Corporation, Armonk, NY, USA). Data were summarised as mean, standard deviation (SD) and range. For statistical calculations, results below the limit of detection (LD; Table S2) were imputed as L D / 2 . The normality of data distribution was assessed using Shapiro–Wilk and Kolmogorov–Smirnov tests. Variables non-normally distributed were Ln transformed and data normality was reassessed. Homogeneity of variance was checked using Levene’s test. For variables with normal distribution, Student’s t-test or Welch’s t-test was used to determine the significance of differences in trace elements concentration between HD patients and the control group. For the variables which remained non-normally distributed after Ln transformation (whole blood Zn and serum Cd), the Mann–Whitney U test was used. Hedges’ g was used to estimate the effect size of chronic HD on trace element concentration. Statistical significance was set at p < 0.05.

3. Results

The mean (SD) age of participants in the whole blood analysis study was 72 (12) years for the HD patient group vs. 64 (19) years for the respective control group; in the serum analysis study it was 69 (14) years for the HD patient group vs. 64 (10) years for the respective control group. The male/female ratio was 61/39 in the HD patient group for whole blood analysis vs. 34/66 in the control group, and 57/43 in the HD patient group for serum analysis vs. 50/50 in the control group.
Statistically significant differences were found between HD patients and controls for all elements (p < 0.05), except for Zn (p = 0.347). Specifically, Li, B, Mn, Ni, Sr, Mo, Cd and Pb concentrations were significantly higher in HD patients (p < 0.05), while Co, Cu, Zn, Se and Rb were significantly lower (p < 0.001; Table 1).
In serum, statistically significant differences were found between HD patients and controls for all elements analysed (p < 0.05). Specifically, Li, B, Co, Ni, Sr, Mo, Cd and Pb concentrations were significantly higher in HD patients (p < 0.05), while Mn, Cu, Zn, Se and Rb concentrations were significantly lower (p < 0.001). The Cu/Zn ratio was also significantly higher in HD patients (p = 0.004; Table 2).

4. Discussion

The results of our study showed a lower concentration of Mn in serum samples from patients in HD compared to a control group of individuals without renal impairment (p < 0.001). In their systematic review with meta-analysis, Tonelli et al. (2009) reported lower Mn concentration in HD patients [21]. More recently, Almeida et al. (2020) also found a significantly lower serum Mn concentration in HD patients, but Stojsavljević et al. (2022) reported increased Mn concentrations [15,34]. It should be noted, as mentioned before, that 96% of circulating Mn is found in erythrocytes, leucocytes and platelets, while only 4% is present in plasma [32]. The small fraction of circulating Mn can be partially lost during the HD process or increased due to contamination of the dialysis fluid [30], which may explain the different results reported in the literature. Furthermore, in our study the mean concentration of Mn in whole blood was significantly higher in HD patients than in the control group (p = 0.025). Two previously published studies also reported the same finding [23,36], while two others found no statistically significant differences [37,38]. Interestingly, one of these latter studies found Mn depositions in the basal ganglia of HD patients, but not in patients who underwent kidney transplantation, patients on peritoneal dialysis or healthy controls [37]. Additionally, despite not having measured Mn, Ninić et al. (2018) found a significantly increased (p = 0.011) gene expression of mitochondrial superoxide dismutase (SOD, a Mn-dependent enzyme) in peripheral blood mononuclear cells of HD patients [39].
Despite the great heterogeneity of results in the literature regarding Cu, Tonelli et al. (2009) reported a higher Cu concentration in HD patients in their meta-analysis [21]. In the present study, we found a significantly decreased concentration of Cu in both whole blood and serum of HD patients compared to the control group (p < 0.001). In a previous study by our group, Almeida et al. (2020) also found a lower serum Cu concentration in a group of HD patients from northern Portugal [34]. Other studies have shown mixed results; Some found no differences in serum [16,18,38] or whole blood Cu concentration [23,36], while others found an increased [15,40,41] or a decreased serum concentration [42]. Copper is mainly excreted in bile [43], which means that loss of renal function is unlikely to significantly interfere with Cu excretion, at least directly. Losses during the HD session [30] and/or low food intake [27] may explain the decreased Cu concentration in chronic HD patients found in some studies. However, oxidative stress and chronic inflammation are two hallmarks of kidney failure and HD [25,26], and Cu is a trace element strongly involved in protection against oxidative stress [43], which may explain the increased concentration of Cu in HD patients found in several studies. Specifically, Cu is a cofactor of the cytoplasmic and extracellular isoforms of SOD, an important enzyme for redox homeostasis [44], whose activity and gene expression have been shown to be increased in erythrocytes [41] and peripheral blood mononuclear cells [39] of HD patients, respectively.
Tonelli et al. (2009) reported significantly decreased Zn concentrations in HD patients in their meta-analysis [21]. This seems to be a consistent finding with regard to serum Zn concentration [15,38,41,42,45], which we also observed both in the present study (p < 0.001), and in a similar study by our group previously published [34]. With regard to the concentration of Zn in whole blood, there are few published studies. For this specimen, we found no statistically significant difference between HD patients and controls (p = 0.347), despite a slightly lower mean Zn concentration in HD patients. Oruc et al. (2022) reported a median concentration (interquartile range) of 3.48 (3.28–3.99) and 3.71 (3.37–3.98) mg/L in HD patients and controls, respectively, with no statistically significant difference (p = 0.240) between groups [36]. On the other hand, Prodanchuk et al. (2013) found a significantly increased mean (SD) concentration of Zn: 5.81 (0.14) mg/L in HD patients vs. 4.93 (0.07) mg/L in the control group (p < 0.001) [23]. However, it is important to note that the Zn concentration in whole blood found in the first study was much lower than in the second and in ours. Zinc balance studies during RRT have provided variable results [30]. Only ~0.1% of total body Zn stores are present in plasma, either weakly bound to albumin (~80%) or strongly bound to α2-macroglobulin (~20%) [46]. Chronic systemic inflammation, which is commonly observed in HD patients [25], may affect serum Zn [24]. In addition to lower dietary intake [27] and potential interactions with Fe supplements [47], all these factors may collectively contribute to the lower serum Zn concentration commonly seen in HD patients.
The serum Cu/Zn ratio was significantly higher (p = 0.004) in HD patients compared to the control group. Inflammation and oxidative stress, two hallmark features of renal failure and chronic HD [25,26], have been shown to influence the serum concentration of both elements, markedly increasing Cu concentration and decreasing Zn concentration [48]. A higher serum Cu/Zn ratio has been associated with an increased risk of all-cause mortality [48], heart failure [49], infection [50], colorectal cancer [51], impaired glomerular filtration rate [52], decreased handgrip strength [53] and impaired glycaemic control [52]. In HD patients, an increased serum Cu/Zn ratio has been shown to be associated with higher levels of total and LDL cholesterol [54] and carotid artery atherosclerosis [40], thus an increased risk of cardiovascular disease.
For Se, we found a significantly lower concentration in both whole blood and serum of HD patients (p < 0.001), as reported by Tonelli et al. (2009) in their meta-analysis [21] and in a previous study by our group [34]. Other recent studies have reported similar results for Se in serum [15,38,41,45]. In whole blood, one study found a decreased concentration of Se in HD patients compared to healthy controls [36], while another study found no statistically significant differences (p = 0.217) [22]. Selenium is another trace element strongly involved in the regulation of the inflammatory process and redox status [55], and its serum concentration seems to be inversely related to the inflammatory status [24]. As mentioned above, increased oxidative stress is commonly observed in patients with renal failure on chronic HD [26]. Several glutathione peroxidase (GPx) isoforms, essential for protection against oxidative damage, are Se-dependent [55]. Some previously published studies observed a significantly decreased GPx activity in HD patients [41,56], but others did not [57]. The causes of this Se deficiency will be several, including dietary restrictions [58]. Additionally, Selenium appears to be lost in the dialysis fluid during the dialysis process [30]. It is important to mention that Se was the only essential trace element that was shown to be strongly and independently associated with an increased risk of death and hospitalisation in a prospective cohort study by Tonelli et al. (2018) [20].
Cobalt’s only known biological function is as a component of vitamin B12 (cobalamin), while other Co compounds are toxic to humans [59]. In this study, we found a significantly decreased whole blood Co concentration (p < 0.001) and a significantly increased serum Co concentration in HD patients (p < 0.001). Only one small study showing no statistically significant differences in Co concentration between HD patients and a control group was included in the review by Tonelli et al. (2009) [21]. In a previous study by our group, an increased serum Co concentration in HD patients was also found [34], while Stojsavlkević et al. (2022) reported a decreased concentration [15]. In whole blood, two previously published studies found a significantly higher (p < 0.001) Co concentration in HD patients [22,36] while in another study the difference was not statistically significant (p = 0.501) [23]. Vitamin B12 and its biologically inactive analogues are found in circulation bound to transcobalamin or haptocorrin [60]. Cellular uptake of vitamin B12 is achieved through the interaction of a cell surface receptor with transcobalamin bound to vitamin B12 (holotranscobalamin) [60]. Holotranscobalamin is filtered by the kidney and is often increased in patients with renal failure [61]. Vitamin B12 does not seem to be lost during HD [30,62], which means that changes in serum Co concentration will result from direct Co losses, contamination of the dialysis fluid due to its contact with Co-containing metallic alloys, or variations in circulating levels of vitamin B12. Despite the increased levels of holotranscobalamin, some studies have shown a lower cellular uptake of vitamin B12 in patients with CKD [63,64], which could explain the lower levels of Co found in whole blood of HD patients.
There are very few studies on Mo status in HD patients. In this study, a significantly increased (p < 0.001) Mo concentration was observed in both whole blood and serum of HD patients. In the meta-analysis by Tonelli et al. (2009), only one study with Mo results was included, which reported increased Mo concentrations in HD patients [21] and Almeida et al. (2020) also found higher serum Mo concentrations in HD patients [34]. Molybdenum is mainly excreted in urine [65], which may explain the increased Mo concentrations observed in HD patients. On the other hand, the dialysis process appears to be able to remove Mo from circulation [66].
Cadmium is a well-established human carcinogen [67]. In this study, we observed a significantly increased Cd concentration in both whole blood and serum of HD patients (p < 0.001). Consistent with our findings, Tonelli et al. (2009) also reported increased Cd levels in HD patients in their meta-analysis [21], as did Almeida et al. (2020) in the serum of a cohort of HD patients from northern Portugal [34]. With only one exception [15], all other recent studies have also reported increased Cd concentrations in both whole blood [22,23,36,68,69,70] and serum [71] of HD patients. In circulation, Cd is mostly found in erythrocytes bound to metallothioneins and other SH-rich proteins and peptides [72,73]. Due to their low molecular weight, serum metallothioneins are filtered and extensively reabsorbed in the kidneys, causing Cd accumulation [74]. As a result, the kidneys constitute the main body reservoir of Cd, with a half-life of several years [75,76]. Loss of renal function impairs the main route of Cd elimination from the circulation [77], which may explain the increased Cd concentrations found in HD patients.
Lead is a toxic trace element with deleterious effects on virtually all functions of the human body [78]. In the present study, a significantly increased concentration of Pb was found in both whole blood and serum of HD patients (p < 0.001). This was also reported by Tonelli et al. (2009) in their meta-analysis [21], and has been confirmed in several other more recent studies, which reported increased Pb levels in both whole blood [22,23,36,38,70,79] and serum [15,34,71] of HD patients. In adult humans, approximately 90% of the total body burden of Pb is found in bone [80]. Mineral bone disorders are often seen in patients with renal failure, mainly due to their advanced age, abnormal metabolism of calcium, phosphate, vitamin D and parathyroid hormone, soft tissue calcification and abnormal bone remodelling and mineralisation [81,82]. In postmenopausal women, bone mineral density progressively decreases while circulating Pb levels increase [83]. It is possible that the increased Pb levels found in HD patients are due to Pb mobilisation during bone resorption caused by an altered bone metabolism.
Serum and whole blood concentrations of Li, B, Ni and Sr were all significantly increased in HD patients (p < 0.05) compared to controls. In the meta-analysis by Tonelli et al. (2009) mixed results were reported for B and Ni, and there is no reference to studies on Li or Sr [21]. In the previous study by our group, an increased serum concentration of Ni and Sr in HD patients was also reported [34]. Other recent studies have reported an increased Li and B concentration in whole blood [23,36] and an increased Ni and Sr concentration in both whole blood [23,36] and serum [15,84], as observed in the present study. All of these trace elements are excreted mainly in the urine [85,86,87,88]. Therefore, the loss of renal function may explain the increased concentrations of Li, B, Ni and Sr observed in HD patients. Additionally, most of the body content of Sr is present in the bone [89]. Mineral bone disturbances, commonly seen in patients with renal failure, as mentioned above, may cause mobilisation of bone Sr (as described for Pb) and explain the increased levels of circulating Sr observed in HD patients. The same might be true for B because it also accumulates in bone [90].
Lastly, we found a significantly decreased concentration of Rb in both whole blood and serum of HD patients (p < 0.001). Data on changes in Rb concentrations in HD patients are very scarce. Almeida et al. (2020) and Stojsavlkević et al. (2022) both reported decreased serum concentrations in HD patients compared to a healthy control group [15,34]. Rubidium appears to be lost in the dialysis fluid during the dialysis process [66,91] and patients with renal failure also appear to have a decreased intake of Rb [92]. Together, these factors may explain the decreased Rb concentrations in HD patients.
Overall, the results of this study show that chronic HD appears to affect the relative distribution of various trace elements between plasma and the intracellular compartment of blood cells. While the concentration of some trace elements appears decreased (e.g., Se) or increased (e.g., Mo) in both whole blood and serum of HD patients, others show opposite variations in whole blood and serum (e.g., Co and Mn). The whole blood/serum concentration ratio of trace elements studied differed between HD patients and controls. The ratios were higher in HD patients than in controls for Pb (350 vs. only 220), Mn (21 vs. 15) and Zn (12 vs. 8.5). In contrast, ratios were lower in HD patients compared to controls for Co (0.8 vs. 1.7) and Mo (0.8 vs. 1.3).
This study has some limitations. First, it does not include data on the aetiology of CKD, which may significantly affect circulating trace element concentrations [54]. Second, the study did not collect information on the duration of chronic HD, which can also affect circulating trace element concentrations [93]. Third, data on dietary intake of the trace elements analysed were not collected. Studies investigating the effect of dietary Zn supplementation on serum Zn concentration in HD patients have produced conflicting results [94,95]. Furthermore, due to dietary restrictions and uremic anorexia associated with CKD and HD [27,28,29], it is plausible that the dietary intake of trace elements is lower in HD patients. Fourth, CKD is usually accompanied by various comorbidities such as diabetes, hypertension and cardiovascular diseases [2,3] that may influence the concentration of trace elements in blood, but no information was collected on comorbidities of HD patients. Lastly, anaemia is a common condition among HD patients [96,97]. Since most trace elements are not evenly distributed between plasma and blood cells, a lower haematocrit affects their concentration in whole blood. These issues should be addressed in future studies.

5. Conclusions

This study confirms that HD patients tend to present significant trace elements imbalances compared to individuals with normal renal function. By determining the concentration of a wide panel of trace elements in both serum and whole blood, it was shown that chronic HD can affect intra- and extracellular blood compartments differently. Thus, while the Se concentration is decreased in both whole blood and serum of HD patients, Co is increased in serum but decreased in whole blood, and Mn is decreased in serum but increased in whole blood. Future studies should further investigate the mechanisms involved in trace element imbalances found in HD patients, including eventual changes at the subcellular level.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/nu15081912/s1, Table S1: Whole blood and serum analysis—quality control data; Table S2: Limits of detection.

Author Contributions

Conceptualisation, A.A.; methodology, D.G., E.P. and A.A.; validation, D.G., E.P. and A.A.; formal analysis, R.A. and A.A.; investigation, D.G. and A.A.; resources, M.D. and A.A.; data curation, R.A. and A.A.; writing—original draft preparation, R.A.; writing—review and editing, R.A. and A.A.; visualisation, R.A.; supervision, A.A.; project administration, A.A.; funding acquisition, A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work received financial support from PT national funds (FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior) through the projects UIDB/50006/2020 and UIDP/50006/2020.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Faculty of Pharmacy of the University of Porto (Parecer Nº 38-06-2019).

Informed Consent Statement

Patient consent was waived due to data anonymization, ensured by removing all patient information except sex and age.

Data Availability Statement

All data related to this study are presented in the article or supplementary material.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kidney Disease Improving Global Outcomes (KDIGO). KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 2013, 3, 1–150. [Google Scholar]
  2. Kovesdy, C.P. Epidemiology of chronic kidney disease: An update 2022. Kidney Int. Suppl. 2022, 12, 7–11. [Google Scholar] [CrossRef]
  3. Sundström, J.; Bodegard, J.; Bollmann, A.; Vervloet, M.G.; Mark, P.B.; Karasik, A.; Taveira-Gomes, T.; Botana, M.; Birkeland, K.I.; Thuresson, M.; et al. Prevalence, outcomes, and cost of chronic kidney disease in a contemporary population of 2.4 million patients from 11 countries: The CaReMe CKD study. Lancet Reg. Health Eur. 2022, 20, 100438. [Google Scholar] [CrossRef] [PubMed]
  4. Liyanage, T.; Ninomiya, T.; Jha, V.; Neal, B.; Patrice, H.M.; Okpechi, I.; Zhao, M.H.; Lv, J.; Garg, A.X.; Knight, J.; et al. Worldwide access to treatment for end-stage kidney disease: A systematic review. Lancet 2015, 385, 1975–1982. [Google Scholar] [CrossRef] [PubMed]
  5. Thurlow, J.S.; Joshi, M.; Yan, G.; Norris, K.C.; Agodoa, L.Y.; Yuan, C.M.; Nee, R. Global Epidemiology of End-Stage Kidney Disease and Disparities in Kidney Replacement Therapy. Am. J. Nephrol. 2021, 52, 98–107. [Google Scholar] [CrossRef] [PubMed]
  6. Htay, H.; Bello, A.K.; Levin, A.; Lunney, M.; Osman, M.A.; Ye, F.; Ashuntantang, G.E.; Bellorin-Font, E.; Gharbi, M.B.; Davison, S.N.; et al. Hemodialysis Use and Practice Patterns: An International Survey Study. Am. J. Kidney Dis. 2021, 77, 326–335. [Google Scholar] [CrossRef]
  7. Baker, L.A.; March, D.S.; Wilkinson, T.J.; Billany, R.E.; Bishop, N.C.; Castle, E.M.; Chilcot, J.; Davies, M.D.; Graham-Brown, M.P.M.; Greenwood, S.A.; et al. Clinical practice guideline exercise and lifestyle in chronic kidney disease. BMC Nephrol. 2022, 23, 75. [Google Scholar] [CrossRef] [PubMed]
  8. Ikizler, T.A.; Burrowes, J.D.; Byham-Gray, L.D.; Campbell, K.L.; Carrero, J.-J.; Chan, W.; Fouque, D.; Friedman, A.N.; Ghaddar, S.; Goldstein-Fuchs, D.J.; et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am. J. Kidney Dis. 2020, 76, S1–S107. [Google Scholar] [CrossRef]
  9. de Rooij, E.N.M.; Meuleman, Y.; de Fijter, J.W.; Le Cessie, S.; Jager, K.J.; Chesnaye, N.C.; Evans, M.; Pagels, A.A.; Caskey, F.J.; Torino, C.; et al. Quality of Life before and after the Start of Dialysis in Older Patients. Clin. J. Am. Soc. Nephrol. 2022, 17, 1159. [Google Scholar] [CrossRef]
  10. Nagasawa, H.; Sugita, I.; Tachi, T.; Esaki, H.; Yoshida, A.; Kanematsu, Y.; Noguchi, Y.; Kobayashi, Y.; Ichikawa, E.; Tsuchiya, T.; et al. The Relationship Between Dialysis Patients’ Quality of Life and Caregivers’ Quality of Life. Front. Pharmacol. 2018, 9, 770. [Google Scholar] [CrossRef]
  11. Nataatmadja, M.; Krishnasamy, R.; Zuo, L.; Hong, D.; Smyth, B.; Jun, M.; de Zoysa, J.R.; Howard, K.; Wang, J.; Lu, C.; et al. Quality of Life in Caregivers of Patients Randomized to Standard-Versus Extended-Hours Hemodialysis. Kidney Int. Rep. 2021, 6, 1058–1065. [Google Scholar] [CrossRef]
  12. Çelik, G.; Annagur, B.B.; Yılmaz, M.; Demir, T.; Kara, F. Are sleep and life quality of family caregivers affected as much as those of hemodialysis patients? Gen. Hosp. Psychiatry 2012, 34, 518–524. [Google Scholar] [CrossRef]
  13. Ayman, K. Advances in Hemodialysis Techniques. In Hemodialysis; Hiromichi, S., Ed.; IntechOpen: Rijeka, Croatia, 2013. [Google Scholar]
  14. Fleming, G.M. Renal replacement therapy review. Organogenesis 2011, 7, 2–12. [Google Scholar] [CrossRef]
  15. Stojsavljević, A.; Ristić-Medić, D.; Krstić, Đ.; Rovčanin, B.; Radjen, S.; Terzić, B.; Manojlović, D. Circulatory Imbalance of Essential and Toxic Trace Elements in Pre-dialysis and Hemodialysis Patients. Biol. Trace Elem. Res. 2022, 200, 3117–3125. [Google Scholar] [CrossRef]
  16. Kiziltas, H.; Ekin, S.; Erkoc, R. Trace element status of chronic renal patients undergoing hemodialysis. Biol. Trace Elem. Res. 2008, 124, 103–109. [Google Scholar] [CrossRef] [PubMed]
  17. Hasanato, R.M. Assessment of trace elements in sera of patients undergoing renal dialysis. Saudi Med. J. 2014, 35, 365–370. [Google Scholar]
  18. Koca, T.; Berber, A.; Koca, H.B.; Demir, T.A.; Koken, T. Effects of hemodialysis period on levels of blood trace elements and oxidative stress. Clin. Exp. Nephrol. 2010, 14, 463–468. [Google Scholar] [CrossRef] [PubMed]
  19. Hossain, M.S.; Amin, M.N.; Das, A.; Khan, A.K.M.J.H.; Sohel, M.; Ahmed, J.; Islam, M.M.; Hossain, M.S.; Rahman, M.M.; Nesa, M.L.; et al. Increased lipid peroxidation, depleted non-enzymatic antioxidant, and variability in trace elements concentration in serum are correlated with Bangladeshi end-stage renal disease population. Health Sci. Rep. 2021, 4, e348. [Google Scholar] [CrossRef]
  20. Tonelli, M.; Wiebe, N.; Bello, A.; Field, C.J.; Gill, J.S.; Hemmelgarn, B.R.; Holmes, D.T.; Jindal, K.; Klarenbach, S.W.; Manns, B.J.; et al. Concentrations of Trace Elements in Hemodialysis Patients: A Prospective Cohort Study. Am. J. Kidney Dis. 2017, 70, 696–704. [Google Scholar] [CrossRef] [PubMed]
  21. Tonelli, M.; Wiebe, N.; Hemmelgarn, B.; Klarenbach, S.; Field, C.; Manns, B.; Thadhani, R.; Gill, J. Trace elements in hemodialysis patients: A systematic review and meta-analysis. BMC Med. 2009, 7, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Shanmugam, L.; Green, S.R.; Radhakrishnan, H.; Kadavanu, T.M.; Ramachandrappa, A.; Tiwari, S.R.; Rajkumar, A.L.; Govindasamy, E. Trace elements in chronic haemodialysis patients and healthy individuals-A comparative study. J. Clin. Diagn. Res. 2016, 10, OC14–OC17. [Google Scholar] [CrossRef]
  23. Prodanchuk, M.; Makarov, O.; Pisarev, E.; Sheiman, B.; Kulyzkiy, M. Disturbances of trace element metabolism in ESRD patients receiving hemodialysis and hemodiafiltration. Cent. Eur. J. Urol. 2013, 66, 472–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Duncan, A.; Talwar, D.; McMillan, D.C.; Stefanowicz, F.; O’Reilly, D.S.J. Quantitative data on the magnitude of the systemic inflammatory response and its effect on micronutrient status based on plasma measurements. Am. J. Clin. Nutr. 2012, 95, 64–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cobo, G.; Lindholm, B.; Stenvinkel, P. Chronic inflammation in end-stage renal disease and dialysis. Nephrol. Dial. Transplant. 2018, 33, iii35–iii40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Liakopoulos, V.; Roumeliotis, S.; Gorny, X.; Dounousi, E.; Mertens, P.R. Oxidative Stress in Hemodialysis Patients: A Review of the Literature. Oxid. Med. Cell. Longev. 2017, 2017, 3081856. [Google Scholar] [CrossRef]
  27. Iorember, F.M. Malnutrition in Chronic Kidney Disease. Front. Pediatr. 2018, 6, 161. [Google Scholar] [CrossRef] [Green Version]
  28. Aguilera, A.; Selgas, R.; Diéz, J.J.; Bajo, M.A.; Codoceo, R.; Alvarez, V. Anorexia in end-stage renal disease: Pathophysiology and treatment. Expert Opin. Pharmacother. 2001, 2, 1825–1838. [Google Scholar] [CrossRef]
  29. Bossola, M.; Di Stasio, E.; Viola, A.; Leo, A.; Carlomagno, G.; Monteburini, T.; Cenerelli, S.; Santarelli, S.; Boggi, R.; Miggiano, G.; et al. Dietary intake of trace elements, minerals, and vitamins of patients on chronic hemodialysis. Int. Urol. Nephrol. 2014, 46, 809–815. [Google Scholar] [CrossRef]
  30. Berger, M.M.; Broman, M.; Forni, L.; Ostermann, M.; De Waele, E.; Wischmeyer, P.E. Nutrients and micronutrients at risk during renal replacement therapy: A scoping review. Curr. Opin. Crit. Care 2021, 27, 367–377. [Google Scholar] [CrossRef]
  31. Covic, A.; Gusbeth-Tatomir, P. Trace elements in end-stage renal disease—Unfamiliar territory to be revealed. BMC Nephrol. 2009, 10, 12. [Google Scholar] [CrossRef] [Green Version]
  32. Milne, D.B.; Sims, R.L.; Ralston, N.V. Manganese content of the cellular components of blood. Clin. Chem. 1990, 36, 450–452. [Google Scholar] [CrossRef] [PubMed]
  33. deSilva, P.E. Determination of lead in plasma and studies on its relationship to lead in erythrocytes. Br. J. Ind. Med. 1981, 38, 209–217. [Google Scholar] [CrossRef] [Green Version]
  34. 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. [Google Scholar] [CrossRef] [PubMed]
  35. Goullé, J.-P.; Mahieu, L.; Castermant, J.; Neveu, N.; Bonneau, L.; Lainé, G.; Bouige, D.; Lacroix, C. Metal and metalloid multi-elementary ICP-MS validation in whole blood, plasma, urine and hair: Reference values. Forensic Sci. Int. 2005, 153, 39–44. [Google Scholar] [CrossRef] [PubMed]
  36. Oruc, M.; Mercan, S.; Bakan, S.; Kose, S.; Ikitimur, B.; Trabulus, S.; Altiparmak, M.R. Do trace elements play a role in coronary artery calcification in hemodialysis patients? Int. Urol. Nephrol. 2022, 55, 173–182. [Google Scholar] [CrossRef]
  37. Akcan, E.; Özkurt, S.; Sahin, G.; Yalcin, A.U.; Adapinar, B. The relation between brain MRI findings and blood manganese levels in renal transplantation, hemodialysis, and peritoneal dialysis patients. Int. Urol. Nephrol. 2018, 50, 173–177. [Google Scholar] [CrossRef]
  38. Gómez de Oña, C.; Martínez-Morillo, E.; Gago González, E.; Vidau Argüelles, P.; Fernández Merayo, C.; Álvarez Menéndez, F.V. Variation of trace element concentrations in patients undergoing hemodialysis in the north of Spain. Scand. J. Clin. Lab. Invest. 2016, 76, 492–499. [Google Scholar] [CrossRef]
  39. Ninić, A.; Sopić, M.; Munjas, J.; Spasojević-Kalimanovska, V.; Kotur-Stevuljević, J.; Bogavac-Stanojević, N.; Ivanišević, J.; Simić-Ogrizović, S.; Kravljača, M.; Jelić-Ivanović, Z. Association Between Superoxide Dismutase Isoenzyme Gene Expression and Total Antioxidant Status in Patients with an End-Stage Renal Disease. Balk. Med. J. 2018, 35, 431–436. [Google Scholar] [CrossRef]
  40. Ari, E.; Kaya, Y.; Demir, H.; Asicioglu, E.; Keskin, S. The correlation of serum trace elements and heavy metals with carotid artery atherosclerosis in maintenance hemodialysis patients. Biol. Trace Elem. Res. 2011, 144, 351–359. [Google Scholar] [CrossRef]
  41. Guo, C.H.; Wang, C.L.; Chen, P.C.; Yang, T.C. Linkage of some trace elements, peripheral blood lymphocytes, inflammation, and oxidative stress in patients undergoing either hemodialysis or peritoneal dialysis. Perit. Dial. Int. 2011, 31, 583–591. [Google Scholar] [CrossRef]
  42. Bhogade, R.B.; Suryakar, A.N.; Joshi, N.G. Effect of hemodialysis on serum copper and zinc levels in renal failure patients. Eur. J. Gen. Med. 2013, 10, 154–157. [Google Scholar] [CrossRef] [PubMed]
  43. Balsano, C.; Porcu, C.; Sideri, S. Is copper a new target to counteract the progression of chronic diseases? Metallomics 2018, 10, 1712–1722. [Google Scholar] [CrossRef] [Green Version]
  44. Damiano, S.; Sozio, C.; La Rosa, G.; Guida, B.; Faraonio, R.; Santillo, M.; Mondola, P. Metabolism Regulation and Redox State: Insight into the Role of Superoxide Dismutase 1. Int. J. Mol. Sci. 2020, 21, 6606. [Google Scholar] [CrossRef]
  45. Kalousová, M.; Kuběna, A.A.; Koštířová, M.; Vinglerová, M.; Ing, O.M.; Dusilová-Sulková, S.; Tesař, V.; Zima, T. Lower retinol levels as an independent predictor of mortality in long-term hemodialysis patients: A prospective observational cohort study. Am. J. Kidney Dis. 2010, 56, 513–521. [Google Scholar] [CrossRef] [PubMed]
  46. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Batchelor, E.K.; Kapitsinou, P.; Pergola, P.E.; Kovesdy, C.P.; Jalal, D.I. Iron Deficiency in Chronic Kidney Disease: Updates on Pathophysiology, Diagnosis, and Treatment. J. Am. Soc. Nephrol. 2020, 31, 456–468. [Google Scholar] [CrossRef] [PubMed]
  48. Malavolta, M.; Piacenza, F.; Basso, A.; Giacconi, R.; Costarelli, L.; Mocchegiani, E. Serum copper to zinc ratio: Relationship with aging and health status. Mech. Ageing Dev. 2015, 151, 93–100. [Google Scholar] [CrossRef] [PubMed]
  49. Kunutsor, S.K.; Voutilainen, A.; Kurl, S.; Laukkanen, J.A. Serum copper-to-zinc ratio is associated with heart failure and improves risk prediction in middle-aged and older Caucasian men: A prospective study. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1924–1935. [Google Scholar] [CrossRef]
  50. Laine, J.T.; Tuomainen, T.-P.; Salonen, J.T.; Virtanen, J.K. Serum copper-to-zinc-ratio and risk of incident infection in men: The Kuopio Ischaemic Heart Disease Risk Factor Study. Eur. J. Epidemiol. 2020, 35, 1149–1156. [Google Scholar] [CrossRef]
  51. Stepien, M.; Jenab, M.; Freisling, H.; Becker, N.-P.; Czuban, M.; Tjønneland, A.; Olsen, A.; Overvad, K.; Boutron-Ruault, M.-C.; Mancini, F.R.; et al. Pre-diagnostic copper and zinc biomarkers and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition cohort. Carcinogenesis 2017, 38, 699–707. [Google Scholar] [CrossRef] [Green Version]
  52. Hamasaki, H.; Kawashima, Y.; Yanai, H. Serum Zn/Cu Ratio Is Associated with Renal Function, Glycemic Control, and Metabolic Parameters in Japanese Patients with and without Type 2 Diabetes: A Cross-sectional Study. Front. Endocrinol. 2016, 7, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Luo, L.; Xu, J.; Jiang, R.; Yao, B.; Di, J. Association between serum copper, zinc and their ratio and handgrip strength among adults: A study from National Health and Nutrition Examination Survey (NHANES) 2011–2014. Environ. Sci. Pollut. Res. Int. 2023, 30, 29100–29109. [Google Scholar] [CrossRef] [PubMed]
  54. Reina de la Torre, M.L.; Navarro-Alarcón, M.; del Moral, L.M.; López, G.d.l.S.H.; Palomares-Bayo, M.; Oliveras López, M.J.; Blanca Herrera, R.M.; Agil, A. Serum Zn levels and Cu/Zn ratios worsen in hemodialysis patients, implying increased cardiovascular risk: A 2-year longitudinal study. Biol. Trace Elem. Res. 2014, 158, 129–135. [Google Scholar] [CrossRef] [PubMed]
  55. Huang, Z.; Rose, A.H.; Hoffmann, P.R. The role of selenium in inflammation and immunity: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2012, 16, 705–743. [Google Scholar] [CrossRef] [Green Version]
  56. Zargari, M.; Sedighi, O. Influence of Hemodialysis on Lipid Peroxidation, Enzymatic and Non-Enzymatic Antioxidant Capacity in Chronic Renal Failure Patients. Nephrourol. Mon. 2015, 7, e28526. [Google Scholar] [CrossRef] [Green Version]
  57. Ozden, M.; Maral, H.; Akaydın, D.; Cetınalp, P.; Kalender, B. Erythrocyte glutathione peroxidase activity, plasma malondialdehyde and erythrocyte glutathione levels in hemodialysis and CAPD patients. Clin. Biochem. 2002, 35, 269–273. [Google Scholar] [CrossRef]
  58. Xie, C.; Zeng, M.; Shi, Z.; Li, S.; Jiang, K.; Zhao, Y. Association between Selenium Status and Chronic Kidney Disease in Middle-Aged and Older Chinese Based on CHNS Data. Nutrients 2022, 14, 2695. [Google Scholar] [CrossRef]
  59. Leyssens, L.; Vinck, B.; Van Der Straeten, C.; Wuyts, F.; Maes, L. Cobalt toxicity in humans—A review of the potential sources and systemic health effects. Toxicology 2017, 387, 43–56. [Google Scholar] [CrossRef]
  60. Sobczyńska-Malefora, A.; Delvin, E.; McCaddon, A.; Ahmadi, K.R.; Harrington, D.J. Vitamin B12 status in health and disease: A critical review. Diagnosis of deficiency and insufficiency—Clinical and laboratory pitfalls. Crit. Rev. Clin. Lab. Sci. 2021, 58, 399–429. [Google Scholar] [CrossRef]
  61. Nexo, E.; Hoffmann-Lücke, E. Holotranscobalamin, a marker of vitamin B-12 status: Analytical aspects and clinical utility. Am. J. Clin. Nutr. 2011, 94, 359s–365s. [Google Scholar] [CrossRef] [Green Version]
  62. Bévier, A.; Novel-Catin, E.; Blond, E.; Pelletier, S.; Parant, F.; Koppe, L.; Fouque, D. Water-Soluble Vitamins and Trace Elements Losses during On-Line Hemodiafiltration. Nutrients 2022, 14, 3454. [Google Scholar] [CrossRef]
  63. Herrmann, W.; Obeid, R.; Schorr, H.; Geisel, J. Functional vitamin B12 deficiency and determination of holotranscobalamin in populations at risk. Clin. Chem. Lab. Med. 2003, 41, 1478–1488. [Google Scholar] [CrossRef] [PubMed]
  64. Obeid, R.; Kuhlmann, M.; Kirsch, C.M.; Herrmann, W. Cellular Uptake of Vitamin B12 in Patients with Chronic Renal Failure. Nephron Clin. Pract. 2005, 99, c42–c48. [Google Scholar] [CrossRef] [PubMed]
  65. Turnlund, J.R.; Keyes, W.R.; Peiffer, G.L. Molybdenum absorption, excretion, and retention studied with stable isotopes in young men at five intakes of dietary molybdenum. Am. J. Clin. Nutr. 1995, 62, 790–796. [Google Scholar] [CrossRef] [PubMed]
  66. Broman, M.; Bryland, A.; Carlsson, O. Trace elements in patients on continuous renal replacement therapy. Acta Anaesthesiol. Scand. 2017, 61, 650–659. [Google Scholar] [CrossRef]
  67. Hartwig, A. Cadmium and cancer. Met. Ions Life Sci. 2013, 11, 491–507. [Google Scholar] [CrossRef]
  68. Palaneeswari, M.S.; Sam Rajan, P.M.A.; Silambanan, S.; Jothimalar. Blood arsenic and cadmium concentrations in end-stage renal disease patients who were on maintenance haemodialysis. J. Clin. Diagn. Res. 2013, 7, 809–813. [Google Scholar] [CrossRef]
  69. Sivrikaya, A.; Menevşe, E.; Altintepe, L.; Tiftik, A.M. The Relations between Levels of Cadmium and Thyroid Parameters in Hemodialysis Patients. J. Clin. Anal. Med. 2013, 4, 1–4. [Google Scholar] [CrossRef]
  70. Chen, B.; Lamberts, L.V.; Behets, G.J.; Zhao, T.; Zhou, M.; Hou, X.; Guan, G.; D’Haese, P.C.; Liu, G. Selenium, lead, and cadmium levels in renal failure patients in China. Biol. Trace Elem. Res. 2009, 131, 1–12. [Google Scholar] [CrossRef]
  71. Kaya, Y.; Ari, E.; Demir, H.; Gecit, I.; Beytur, A.; Kaspar, C. Serum cadmium levels are independently associated with endothelial function in hemodialysis patients. Int. Urol. Nephrol. 2012, 44, 1487–1492. [Google Scholar] [CrossRef]
  72. Nordberg, G.F.; Piscator, M.; Nordberg, M. On the distribution of cadmium in blood. Acta Pharmacol. Toxicol. 1971, 30, 289–295. [Google Scholar] [CrossRef] [PubMed]
  73. Zalups, R.K.; Ahmad, S. Molecular handling of cadmium in transporting epithelia. Toxicol. Appl. Pharmacol. 2003, 186, 163–188. [Google Scholar] [CrossRef] [PubMed]
  74. Nordberg, M.; Nordberg, G.F. Toxicological aspects of metallothionein. Cell. Mol. Biol. 2000, 46, 451–463. [Google Scholar] [PubMed]
  75. Nordberg, G.F.; Nogawa, K.; Nordberg, M.; Friberg, L.T. Cadmium. In Handbook on the Toxicology of Metals, 3rd ed.; Nordberg, G.F., Fowler, B.A., Nordberg, M., Friberg, L.T., Eds.; Academic Press: Burlington, ON, Canada, 2007; pp. 445–486. [Google Scholar]
  76. Nordberg, G.F.; Kjellstrom, T.; Nordberg, M. Kinetics and metabolism. In Cd and Health: A Toxicological and Epidemiological Appraisal. Vol I: Exposure, Dose and Metabolism, 1st ed.; Friberg, L.T., Carl-Gustaf, E., Kjellstrom, T., Nordberg, G.F., Eds.; CRC Press: Boca Raton, FL, USA, 1985; pp. 103–178. [Google Scholar]
  77. Godt, J.; Scheidig, F.; Grosse-Siestrup, C.; Esche, V.; Brandenburg, P.; Reich, A.; Groneberg, D.A. The toxicity of cadmium and resulting hazards for human health. J. Occup. Med. Toxicol. 2006, 1, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Wani, A.L.; Ara, A.; Usmani, J.A. Lead toxicity: A review. Interdiscip. Toxicol. 2015, 8, 55–64. [Google Scholar] [CrossRef] [Green Version]
  79. Subha Palaneeswari, M.; Abraham Sam Rajan, P.M.; Silambanan, S.; Jothimalar. Blood lead in end-stage renal disease (ESRD) patients who were on maintainence haemodialysis. J. Clin. Diagn. Res. 2012, 6, 1633–1635. [Google Scholar] [CrossRef]
  80. Flora, G.; Gupta, D.; Tiwari, A. Toxicity of lead: A review with recent updates. Interdiscip. Toxicol. 2012, 5, 47–58. [Google Scholar] [CrossRef]
  81. Tasnim, N.; Dutta, P.; Nayeem, J.; Masud, P.; Ferdousi, A.; Ghosh, A.S.; Hossain, M.; Rajia, S.; Kubra, K.T.; Sakibuzzaman, M.; et al. Osteoporosis, an Inevitable Circumstance of Chronic Kidney Disease: A Systematic Review. Cureus 2021, 13, e18488. [Google Scholar] [CrossRef]
  82. Evenepoel, P.; Cunningham, J.; Ferrari, S.; Haarhaus, M.; Javaid, M.K.; Lafage-Proust, M.-H.; Prieto-Alhambra, D.; Torres, P.U.; Cannata-Andia, J.; European Renal Osteodystrophy workgroup; et al. European Consensus Statement on the diagnosis and management of osteoporosis in chronic kidney disease stages G4–G5D. Nephrol. Dial. Transplant. 2020, 36, 42–59. [Google Scholar] [CrossRef]
  83. Silbergeld, E.K.; Schwartz, J.; Mahaffey, K. Lead and osteoporosis: Mobilization of lead from bone in postmenopausal women. Environ. Res. 1988, 47, 79–94. [Google Scholar] [CrossRef]
  84. Brrow, F.; Bargiul, S. Estimation of nickel levels in blood serum among hemodialysis patients in Syria. Res. J. Pharm. Technol. 2021, 14, 1507–1510. [Google Scholar] [CrossRef]
  85. Uluisik, I.; Karakaya, H.C.; Koc, A. The importance of boron in biological systems. J. Trace Elem. Med. Biol. 2018, 45, 156–162. [Google Scholar] [CrossRef] [PubMed]
  86. Timmer, R.T.; Sands, J.M. Lithium Intoxication. J. Am. Soc. Nephrol. 1999, 10, 666–674. [Google Scholar] [CrossRef]
  87. Sunderman, F.W.; Aitio, A.; Morgan, L.G.; Norseth, T. Biological Monitoring of Nickel. Toxicol. Ind. Health 1986, 2, 17–78. [Google Scholar] [CrossRef]
  88. International Programme on Chemical Safety & Inter-Organization Programme for the Sound Management of Chemicals. Strontium and Strontium Compounds; World Health Organization: Geneva, Switzerland, 2010. [Google Scholar]
  89. Kołodziejska, B.; Stępień, N.; Kolmas, J. The Influence of Strontium on Bone Tissue Metabolism and Its Application in Osteoporosis Treatment. Int. J. Mol. Sci. 2021, 22, 6564. [Google Scholar] [CrossRef] [PubMed]
  90. Rondanelli, M.; Faliva, M.A.; Peroni, G.; Infantino, V.; Gasparri, C.; Iannello, G.; Perna, S.; Riva, A.; Petrangolini, G.; Tartara, A. Pivotal role of boron supplementation on bone health: A narrative review. J. Trace Elem. Med. Biol. 2020, 62, 126577. [Google Scholar] [CrossRef]
  91. Scharfetter, H.; Krachler, M.; Wirnsberger, G.H.; Holzer, H.; Hutten, H. Kinetic modeling of rubidium during hemodialysis. In Proceedings of the 19th Annual International Conference of the IEEE Engineering in Medicine and Biology Society. ‘Magnificent Milestones and Emerging Opportunities in Medical Engineering’ (Cat. No.97CH36136), Chicago, IL, USA, 30 October–2 November 1997; Volume 2135, pp. 2135–2137. [Google Scholar]
  92. Canavese, C.; Decostanzi, E.; Bergamo, D.; Sabbioni, E.; Stratta, P. Rubidium, salami and depression. You cannot have everything in life. Blood Purif. 2008, 26, 311–314. [Google Scholar] [CrossRef] [PubMed]
  93. Esfahani, S.T.; Hamidian, M.R.; Madani, A.; Ataei, N.; Mohseni, P.; Roudbari, M.; Haddadi, M. Serum zinc and copper levels in children with chronic renal failure. Pediatr. Nephrol. 2006, 21, 1153–1156. [Google Scholar] [CrossRef]
  94. Guo, C.H.; Wang, C.L. Effects of zinc supplementation on plasma copper/zinc ratios, oxidative stress, and immunological status in hemodialysis patients. Int. J. Med. Sci. 2013, 10, 79–89. [Google Scholar] [CrossRef] [Green Version]
  95. Tonelli, M.; Wiebe, N.; Thompson, S.; Kinniburgh, D.; Klarenbach, S.W.; Walsh, M.; Bello, A.K.; Faruque, L.; Field, C.; Manns, B.J.; et al. Trace element supplementation in hemodialysis patients: A randomized controlled trial. BMC Nephrol. 2015, 16, 52. [Google Scholar] [CrossRef] [Green Version]
  96. Liu, Y.; Tang, R.; Xu, Q.; Xu, S.; Zuo, S.; Qiu, J.; Zhong, X.; Tan, R.; Liu, Y. High Blood Cu/Zn Ratio is Associated with Nutritional Risk in Patients Undergoing Maintenance Hemodialysis. Biol. Trace Elem. Res. 2022, 200, 4977–4987. [Google Scholar] [CrossRef] [PubMed]
  97. Zuo, S.; Liu, M.; Liu, Y.; Xu, S.; Zhong, X.; Qiu, J.; Qin, D.; Tan, R.; Liu, Y. Association Between the Blood Copper-Zinc (Cu/Zn) Ratio and Anemia in Patients Undergoing Maintenance Hemodialysis. Biol. Trace Elem. Res. 2022, 200, 2629–2638. [Google Scholar] [CrossRef] [PubMed]
Table 1. Mean (SD) and [range] for the trace element concentrations (µg/L) in whole blood of haemodialysis patients and controls. Hedges’ g was used to estimate the effect size of haemodialysis on trace element concentration.
Table 1. Mean (SD) and [range] for the trace element concentrations (µg/L) in whole blood of haemodialysis patients and controls. Hedges’ g was used to estimate the effect size of haemodialysis on trace element concentration.
Trace ElementHaemodialysisControlsHedges’ g
(95% CI)
p-Value
Li2.66 (1.92)
[0.55–8.71]
1.07 (0.81)
[0.25–3.43]
1.37 (1.00–1.75)<0.001
B133 (62)
[45–405]
46 (23)
[10–105]
2.31 (1.89–2.71)<0.001
Mn8.4 (2.3)
[4.1–14.9]
7.6 (1.5)
[4.5–11.0]
0.35 (0.02–0.67)0.025
Co0.188 (0.062)
[0.057–0.366]
0.226 (0.049)
[0.146–0.364]
−0.75 (−1.09–−0.40)<0.001
Ni1.82 (0.46)
[0.91–2.98]
1.53 (0.69)
[0.71–3.25]
0.71 (0.38–1.04)<0.001
Cu843 (138)
[592–1200]
955 (167)
[512–1365]
−0.72 (−1.05–−0.39)<0.001
Zn5360 (755)
[3764–7341]
5767 (2249)
[2254–9756]
−0.04 (−0.35–0.28)0.347
Se123 (19)
[75–169]
138 (20)
[92–184]
−0.76 (−1.09–−0.43)<0.001
Rb1142 (221)
[635–1604]
2457 (670)
[1087–3993]
−2.97 (−3.42–−2.51)<0.001
Sr29.9 (4.0)
[21.6–40.4]
17.6 (6.7)
[7.3–35.2]
2.40 (1.99–2.82)<0.001
Mo2.48 (1.32)
[0.07–5.84]
0.80 (0.26)
[0.30–1.50]
1.53 (1.16–1.90)<0.001
Cd1.00 (0.32)
[0.38–1.86]
0.299 (0.136)
[0.094–0.655]
3.31 (2.81–3.81)<0.001
Pb100 (24)
[42–158]
21.1 (14.2)
[5.8–77.8]
4.13 (3.57–4.68)<0.001
Table 2. Mean (SD) and [range] for the trace element concentrations (µg/L) and Cu/Zn ratio in serum of haemodialysis patients and controls. Hedges’ g was used to estimate the effect size of haemodialysis on trace element concentration.
Table 2. Mean (SD) and [range] for the trace element concentrations (µg/L) and Cu/Zn ratio in serum of haemodialysis patients and controls. Hedges’ g was used to estimate the effect size of haemodialysis on trace element concentration.
Trace ElementHaemodialysisControlsHedges’ g
(95% CI)
p-Value
Li2.89 (1.75)
[0.70–7.92]
1.19 (0.76)
[0.25–2.94]
1.56 (0.96–2.15)<0.001
B89 (42)
[22–211]
34 (17)
[15–64]
1.88 (1.28–2.47)<0.001
Mn0.40 (0.10)
[0.22–0.65]
0.52 (0.11)
[0.36–0.76]
−1.16 (−1.72–−0.58) <0.001
Co0.23 (0.11)
[0.12–0.72]
0.130 (0.037)
[0.094–0.214]
1.67 (1.09–2.26)<0.001
Ni2.80 (0.95)
[0.90–5.50]
1.09 (0.25)
[0.78–1.58]
1.90 (1.25–2.53)<0.001
Cu671 (150)
[341–1046]
812 (139)
[621–1070]
−0.93 (−1.49–−0.40)<0.001
Zn448 (84)
[271–658]
681 (114)
[475–876]
−2.59 (−3.23–−1.95) <0.001
Se62 (13)
[32–90]
82 (15)
[58–103]
−1.43 (−1.99–−0.87) <0.001
Rb87 (24)
[34–146]
115 (19)
[89–145]
−1.14 (−1.71–−0.57) <0.001
Sr29.0 (4.4)
[19.7–39.8]
22.3 (7.6)
[10.5–36.0]
1.32 (0.76–1.88)0.003
Mo3.20 (1.48)
[0.16–6.79]
0.61 (0.42)
[0.026–1.35]
1.87 (1.27–2.45)<0.001
Cd0.038 (0.016)
[0.008–0.080]
0.013 (0.005)
[0.008–0.018]
2.64 (1.97–3.27)<0.001
Pb0.287 (0.149)
[0.025–0.755]
0.096 (0.083)
[0.021–0.317]
2.55 (1.88–3.20)<0.001
Cu/Zn1.54 (0.43)
[0.76–2.54]
1.22 (0.25)
[0.77–1.65]
0.80 (0.26–1.34)0.004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Azevedo, R.; Gennaro, D.; Duro, M.; Pinto, E.; Almeida, A. Further Evidence on Trace Element Imbalances in Haemodialysis Patients—Paired Analysis of Blood and Serum Samples. Nutrients 2023, 15, 1912. https://doi.org/10.3390/nu15081912

AMA Style

Azevedo R, Gennaro D, Duro M, Pinto E, Almeida A. Further Evidence on Trace Element Imbalances in Haemodialysis Patients—Paired Analysis of Blood and Serum Samples. Nutrients. 2023; 15(8):1912. https://doi.org/10.3390/nu15081912

Chicago/Turabian Style

Azevedo, Rui, Davide Gennaro, Mary Duro, Edgar Pinto, and Agostinho Almeida. 2023. "Further Evidence on Trace Element Imbalances in Haemodialysis Patients—Paired Analysis of Blood and Serum Samples" Nutrients 15, no. 8: 1912. https://doi.org/10.3390/nu15081912

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