Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Nephrol Dial Transplant (2013) 28: 716–723 doi: 10.1093/ndt/gfs170 Advance Access publication 4 July 2012 Original Articles 1 Moosa Salehi1, Department of Nutrition, Shiraz School of Health and Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran, Zahra Sohrabi1, 2 1 Maryam Ekramzadeh , Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran, 3 2 Mohammad Kazem Fallahzadeh , Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran and Maryam Ayatollahi3, 4 3 Bita Geramizadeh , Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran Jafar Hassanzadeh1 and Mohammad Mahdi Sagheb4 Keywords: anorexia, antioxidants, nutrition assessment, trace elements, uremia Correspondence and offprint requests to: Mohammad Mahdi Sagheb; E-mail: saghebf@gmail.com † The authors declare that the results presented in this paper have not been published previously in whole or part, except in the abstract form. ferritin and transferrin as well as the subjective global assessment (SGA) score, malnutrition–inflammation score (MIS) and hemoglobin (Hb) levels were measured at the baseline and at the end of the treatment phase. The primary outcome was a change in the nutritional status measured by the SGA score from the baseline towards the end of the treatment phase of the study. Results. The SGA score and MIS decreased significantly in the selenium group compared to the placebo group (P < 0.001 for both). Moreover, serum levels of MDA decreased significantly in the selenium group compared with increasing levels in the placebo group (P < 0.001). Selenium supplementation also hindered an increase in IL-6 levels compared with the placebo group (P = 0.016). There were no significant differences between the selenium and placebo groups in terms of changes in serum levels of lipoproteins, HSCRP, homocysteine, ferritin and transferrin or Hb levels. A B S T R AC T Background. Malnutrition is highly prevalent in hemodialysis (HD) patients. These patients have high levels of oxidative stress and inflammation which can subsequently induce malnutrition. Selenium levels have been found to be decreased in HD patients. As selenium deficiency leads to oxidative stress and inflammatory response, the aim of this study was to evaluate the effects of selenium supplementation on oxidative and inflammatory markers and the nutritional status of HD patients. Methods. In this randomized double-blind placebo-controlled trial, 80 patients on stable HD for at least 3 months without any acute illness or active infections were randomly allocated to two equal groups to receive one selenium (200 µg) or placebo capsule daily for 12 weeks. Serum levels of lipoproteins, malondialdehyde (MDA), interleukin-6 (IL-6), high-sensitivity C-reactive protein (HSCRP), homocysteine, © The Author 2012. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved. 716 Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 Selenium supplementation improves the nutritional status of hemodialysis patients: a randomized, double-blind, placebocontrolled trial† INTRODUCTION (i) The patients in the first group received one capsule of selenium in the form of selenium yeast (200 µg) daily for 12 weeks. (ii) The patients in the second group received one capsule of placebo daily for 12 weeks. Blocked randomization with a fixed block size of four was performed by one of the investigators who had no clinical involvement in the study using Random Allocation Software [17]. All selenium and placebo capsules were provided by Shiraz School of Pharmacy in prepacked bottles numbered for each patient according to the randomization sequence. Each patient was assigned to an order number and received the selenium or placebo in the corresponding prepacked bottles. The placebo and selenium capsules were completely similar in size, weight, color and taste. All of the patients, clinical investigators, including the one who administered SGA questionnaires, and other health care staff were blinded to the treatment assignment. Selenium has a narrow window of the therapeutic index. The maximum recommended allowance dose for selenium is 400–450 µg/day. As the baseline selenium levels of the patients were not measured in this study, in order to maintain the patients’ safety and reduce the chance of selenium toxicity, the dose of selenium supplementation was chosen to be 200 µg/day. Moreover, in support of choosing this dose, a 3-month treatment of HD patients with the same dose of oral selenium has been demonstrated to be safe and effective in restoring levels of selenium and in attenuating oxidative DNA damage in a study by Zachara et al. [18]. SGA is a comprehensive nutritional assessment tool that has several strengths: it is inexpensive and rapid to conduct, can be easily implemented in different disciplines, needs no laboratory evaluation and has been shown to be valid and M AT E R I A L S A N D M E T H O D S This randomized, double-blind, placebo-controlled, two-arm parallel trial was done in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines. The Ethics Committee of Shiraz University of Medical Sciences reviewed and approved the protocol of this study. Eligible participants were 18- to 80-year-old patients who were under regular HD 717 Selenium for malnutrition in hemodialysis patients ORIGINAL ARTICLES Malnutrition or insufficient protein-calorie intake is highly prevalent in hemodialysis (HD) patients [1]. Protein-energy wasting and inflammation are concurrent conditions in HD patients and are associated with poor prognosis [1]. Hence, the term malnutrition–inflammation complex syndrome (MICS) was used to comprise both malnutrition and inflammation, regardless of the original causes [1, 2]. MICS is considered as one of the main causes of atherosclerotic cardiovascular disease (CVD), hospitalization and increased mortality in HD patients [1]. Among the contributing factors to the pathogenesis of MICS in HD patients, high oxidative stress is considered to be one of the most important ones [3, 4]. High oxidative stress in HD patients results from increased production of oxidative compounds and impaired antioxidant defense mechanisms [5–7]. Selenium is an essential trace element with known antioxidant properties [8, 9]. It acts as a cofactor for the reduction in important antioxidant enzymes like glutathione peroxidase [10]. Several studies have demonstrated that HD patients have low levels of selenium compared with healthy controls, and deficiency of this trace element may contribute to increased oxidative stress and inflammation in HD patients [10–13]. Furthermore, strong positive correlations between levels of selenium and nutritional markers such as serum albumin have been reported [14, 15]. Therefore, selenium deficiency may also contribute to malnutrition in HD patients [11]. So far, a few small non-controlled trials have shown the efficacy of selenium supplementation in reducing oxidative stress markers in HD patients [6]. However, no clinical study has ever evaluated the efficacy of selenium supplementation on improving clinical outcomes such as malnutrition in these patients. Therefore, we performed this trial to evaluate the efficacy of selenium supplementation on improving the nutritional status of HD patients. Furthermore, to delineate its possible mechanisms of action, we evaluated the effects of selenium supplementation on serum levels of malondialdehyde (MDA) as a surrogate marker for oxidative stress and on serum levels of interleukin-6 (IL-6) and highsensitivity C-reactive protein (HSCRP) as surrogate markers for inflammation. Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 in Faghihi Hospital Hemodialysis Center for 3 months or more. Exclusion criteria were pregnancy, taking antioxidant supplements including vitamin E, vitamin C, lipoic acid, omega-3 fatty acids, soy extracts and green-tea preparations or immunosuppressive medications within 2 months prior to enrollment in the study, hospitalization in the previous month before the onset of the trial, or having active infection. Sample size was determined according to the subjective global assessment (SGA) score in a previous study in malnourished HD patients [16]. A sample size of 25 patients per group was obtained with a mean difference of 0.8, an SD of 1 and a probability of 80% at the predetermined level of α = 0.05. While considering 15 dropouts in each group, the final sample size was determined to be 40 patients per group. After screening all of the 280 patients under regular HD in Faghihi Hospital Hemodialysis Center, 80 stable patients were found to be eligible for enrollment and gave their informed consent to participate in this trial. These participants were dialyzed three times a week by low-flux dialyzer with polysulfone/polyamide membranes, reverse osmosis purified water and bicarbonate-containing dialysate. These patients were randomly assigned in a 1:1 ratio into two groups: Conclusions. This study shows that selenium may be an effective complementary supplement for reducing the severity of malnutrition in HD patients through alleviating oxidative stress and inflammation. ORIGINAL ARTICLES SA, Belgium ELISA kits for IL-6; Biomerica, CA, USA ELISA kits for PTH; Axis Shield, UK ELISA kits for homocysteine; IBL, Germany ELISA kits for HSCRP and ferritin); intra- and interassay coefficients of variation were 4.2 and 4.4%, 2.8 and 5.1%, 7 and 9%, 4.1 and 5.8%, and 7.8 and 7.6%, respectively. Other parameters were measured with standard automated techniques. Adverse events were inspected by clinical staff in each dialysis session during the treatment phase of the trial. Statistical analyses were done using SPSS version 15 (SPSS Inc., Chicago, IL) statistical software package. Normally distributed data were compared between two groups by an independent sample t-test; skewed data were compared by the Mann–Whitney U-test and categorical data such as sex by χ 2 test. P-values <0.05 were considered statistically significant. The primary outcome of this trial was an absolute change in nutritional statuses of patients measured by the SGA score from the baseline to the end of the treatment phase of the study in an intention-to-treat population. This trial is registered with Clinicaltrial.gov, number NCT01147354, where the trial protocol could be accessed. R E S U LT S Eligible participants were recruited from March 2009 to June 2009. As demonstrated in the flow diagram of the study (Figure 1), during the treatment phase of the study, 11 patients were excluded in the selenium group and 4 in the placebo group. The reasons for these exclusions are mentioned in the flow diagram. Baseline demographic and laboratory characteristics of patients in both groups are demonstrated in Table 1; no statistically significant differences were observed between these two groups in terms of baseline characteristics. All of the enrolled patients were under regular treatment with erythropoietin and intravenous iron based on our regional HD guideline. The changes in measured parameters during the treatment phase of the study are mentioned in Table 2. SGA scores and MIS decreased in the selenium group while these scores increased in the placebo group; when changes in these scores were compared between the two groups, decrements in SGA scores and MIS in the selenium group were significantly different from the corresponding increments in the placebo group (P < 0.001 for both). Likewise, a decrement in serum levels of MDA was significantly different from the corresponding increment in the placebo group (P < 0.001). Moreover, serum levels of IL-6 increased in both groups; however, this increment was significantly lower in the selenium group (P = 0.016). Changes in other parameters were not significantly different between two groups. The frequency of serious adverse events and other adverse events in patients is demonstrated in Table 3. Other adverse events occurred in the first 2 weeks of treatment, and none of them was bothersome enough to compel the patients to quit the treatment. 718 M. Salehi et al. Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 reliable in HD patients [19, 20]. Despite these advantages, it should be appreciated that the status of visceral proteins such as albumin is not well assessed by SGA. To compensate for this problem, the malnutrition–inflammation score (MIS) has been developed that incorporates the serum laboratory markers of malnutrition with SGA and is suggested to be a better indicator of MICS [19, 21, 22]. Therefore, in this study, the nutritional statuses of all patients were assessed by both SGA and MIS. At the baseline, the questions of the SGA questionnaire were read to the patients and completed in person by the main investigator of this study who is an experienced nutritionist working with HD patients regularly. The questions of SGA questionnaire were also asked by the same person at the end of the treatment phase. Furthermore, the physical examinations were done by the same person at the baseline and at the end of the treatment phase. The SGA questionnaire inquired about any changes in weight (during the preceding 6 months and 2 weeks), dietary intake, gastrointestinal symptoms, functional capacity and an assumed metabolic demand of the underlying disease. The physical examination consisted of loss of subcutaneous fat, muscle wasting and the presence of ankle/sacral edema. Each of these features were rated as A, B or C separately to indicate the degree of malnutrition. Then, the SGA classifications were converted to numerical equivalents: a score of <10 points was regarded as wellnourished; 10–17 points, at risk for malnutrition or mildly to moderately malnourished; and more than 17 points, severely malnourished [23]. MIS consisted of four sections (nutritional history, physical examination, BMI and laboratory values) and 10 components. The first three sections included five components adopted from the original SGA and the fourth MIS section included two laboratory values: serum albumin and transferrin. Each component had four levels of severity, from 0 (normal) to 3 (severely abnormal). The sum of all 10 MIS components ranged from 0 (normal) to 30 (severely malnourished); a higher score reflected a more severe degree of malnutrition and inflammation [21]. Before the onset of the treatment and after the end of the treatment phase of the study, 10 cc blood samples were taken from each patient. The blood was taken from the patient’s arm used for HD cannulae just before the beginning of the HD session. The serum was separated by centrifugation at 3000 g/min for 5 min and stored at −70°C. Serum levels of MDA (µmol/L), IL-6 ( pg/mL), HSCRP (μg/mL), ferritin (ng/ mL), transferrin (μg/dL), homocysteine (μmol/L), calcium (mg/dL), phosphate (mg/dL), parathyroid hormone (PTH) ( pg/dL), albumin (g/dL), blood urea nitrogen (BUN) (mg/ dL) and creatinine (mg/dL) as well as hemoglobin (Hb) levels (g/dL) were measured in all patients at the baseline and at the end of the treatment phase of the study. Serum concentrations of MDA were measured by the modified thiobarbituric acid method (spectrophotometric method); the intra- and interassay coefficients of variation were 5.5 and 5.9%, respectively [24]. Serum levels of IL-6, PTH, homocysteine, HSCRP and ferritin were measured using highly sensitive ELISA kits (DIAsource Immunoassays Our study, for the first time, demonstrated the efficacy of selenium supplementation in improving the nutritional status of HD patients. This effect is probably mediated by inhibiting oxidative stress and inflammation. High oxidative stress and generalized inflammation are implicated in the pathogenesis of several complications of HD like atherosclerotic CVD, anemia and malnutrition [3, 7, 25]. This high oxidative stress in HD patients results from an imbalance between pro- and antioxidant factors [6, 7]. Among defense mechanisms against oxidative injury, selenium-dependent enzymes like glutathione peroxidase are the most important ones [5, 8, 25]. There are several reports on the deficiency of selenium and its dependent enzymes in HD patients [5, 12, 13, 26]. Furthermore, low levels of selenium in HD patients have been reported to be associated with increased inflammatory markers [27]. There are also reports on a possible association of selenium deficiency with increased risk of malnutrition in HD patients evidenced by significant positive correlations found between low levels of selenium and of nutritional markers such as serum albumin, total muscle mass, triceps skin-fold and mid-arm muscle circumference [11, 14, 15, 26, 28]. Although few interventional studies have demonstrated the beneficial effects of selenium supplementation on reducing oxidative stress markers or increasing the activity of antioxidant enzymes in HD patients [28–32], the potential efficacy of selenium supplementation 719 Selenium for malnutrition in hemodialysis patients ORIGINAL ARTICLES in improving clinical outcomes like malnutrition in these patients has not been evaluated in any randomized controlled trial so far. In our study, a decrease in serum levels of MDA in the selenium group was significantly higher than placebo. In an interventional study by Ardalan et al. [32], selenium and vitamin E administration to HD patients prevented the iron infusion-induced increase in serum levels of MDA. In another interventional study by Koenig et al. [31], selenium supplementation decreased plasma levels of MDA and increased levels of antioxidant enzymes like glutathione peroxidase in HD patients. Furthermore, in a recent interventional study, Zachara et al. [18] have demonstrated that HD patients have higher oxidative DNA damage in white blood cells compared with healthy controls, and selenium supplementation could significantly dampen this damage. Moreover, in our study, an increase in serum levels of IL-6 levels was significantly lower in the selenium group than placebo. By regulating the expression of selenoprotein genes, selenium has been shown to inhibit the activation of nuclear factor kappa B (NF-κB) signaling pathway and therefore decrease the production of inflammatory cytokines such as IL-6 [33, 34]. Therefore, based on the findings of our study, the observed beneficial effect of selenium supplementation on improving the nutritional status of HD patients may be partly mediated by the inhibition of oxidative damage and inflammation. However, in this study, changes in serum levels of other inflammatory markers such as HSCRP were not significantly different between two groups. These DISCUSSION Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 F I G U R E 1 : Flow diagram of the trial. Table 1. Baseline demographic characteristics and measured parameters in patients Demographic characteristics and measured parameters in patients Selenium Placebo P-value Number 40 40 — 17 (42.5)a 17 (42.5) — Diabetic nephropathy 14 (35) 11 (27.5) Renal stone 1 (2.5) 0 (0) Polycystic kidney disease 0 (0) 1 (2.5) Pyelonephritis 2 (5) 1 (2.5) Glomerulonephritis 1 (2.5) 1 (2.5) Unknown 5 (12.5) 9 (22.5) Age (years) 50 ± 15.4 55 ± 13 NS Gender (female) 24 (60) 20 (50) NS BMI (kg/m ) 22.86 ± 4.07 22.37 ± 3.52 NS Daily Kt/V 1.42 ± 0.26 1.34 ± 0.17 NS Creatinine (mg/dL) 7.75 ± 2.6 8.5 ± 4.11 NS BUN (mg/dL) 64.04 ± 21.3 64.9 ± 28.9 NS Calcium (mg/dL) 8.12 ± 1.1 8.13 ± 0.89 NS Phosphorus (mg/dL) 5.25 ± 1.8 5.64 ± 1.6 NS PTH (pg/dL) 84.9 (8.85, 318.97) 96.7 (25.97, 221.6) NS Albumin (g/dL) 4.43 ± 0.69 4.1 ± 1.06 NS SGA score 12 (9, 18) 11 (8.25, 12) NS MIS 9 (4.25, 14.75) 8 (5, 10) NS MDA (µmol/L) 11.83 ± 3.02 11.56 ± 2.9 NS IL-6 (pg/mL) 33.55 (8.37, 53.45) 14.55 (0.5, 53.17) NS HSCRP (μg/mL) 4.5 (1.72,19.97) 6.7 (2.75, 26.77) NS TG (mg/dL) 104.53 ± 58.59 121.5 ± 60.73 NS Total cholesterol (mg/dL) 155.81 ± 33.77 163.5 ± 39.31 NS LDL (mg/dL) 88.09 ± 27.88 89.45 ± 34.3 NS HDL (mg/dL) 46.08 ± 17.1 49.75 ± 14.21 NS Homocysteine (μmol/L) 23.7 ± 10.38 24.45 ± 8.25 NS Ferritin (ng/mL) 819.4 (439.95, 1180) 838.9 (468.52, 1048.5) NS Total iron binding capacity (μg/dL) 315 (204, 454) 303 (147, 474.75) NS Hb (g/dL) 9.94 ± 2.28 10.4 ± 2.22 NS Causes of ESRD (frequency) Hypertensive nephropathy NS, not significant. Categorical data are expressed as number (%), continuous data with normal distribution as means ± SDs and continuous data with skewed distribution as median (IQR). a differences may reach significance levels in studies with larger sample sizes and longer durations. In addition to its antioxidant and anti-inflammatory properties, the observed beneficial effects of selenium on the nutritional status may be attributed to its effects on the digestion and absorption of fat, nutrient utilization, decreasing ketone bodies (as appetite suppressants) and improving insulin effects [35–38]. 720 M. Salehi et al. ORIGINAL ARTICLES Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 2 Table 2. Effect of selenium supplementation on levels of measured parameters in patients Placebo group P-value SGA score −3.89 ± 3.2 [−5.13 to −2.65]a 1.35 ± 3.01 [0.3–2.4] <0.001 MIS −4.17 ± 4.2 [−5.81 to −2.54] 0.7 ± 3.71 [−0.58 to 2] <0.001 MDA (µmol/l) −1.2 (−3.17, −0.45) [−2.41 to −1.16] 0.6 (0.01, 2.22) [0.47–1.86] <0.001 IL-6 (pg/mL) 6.05 (−20.4, 50.8) [−61.4 to 89.68] 22.95 (0.92, 1978.2) [336.33–1218.3] 0.016 HSCRP (μg/mL) −0.85 (−2.47, 5.25) [−1.99 to 16.46] 1.3 (−17.7, 4.52) [−13.69 to 5.08] NS TG (mg/dL) 14.5 (−2.5, 54.25) [3.06– 50.55] 23 (−12, 54) [−7.25 to 36.28] NS Total cholesterol(mg/dL) −13.7 ± 50.4 [−34.09 to 6.63] −8.02 ± 59.6 [−28.51 to 12.45] NS LDL (mg/dL) −15.2 ± 45.27 [−33.5 to 3.07] −5.44 ± 53.86 [−23.94 to 13.06] NS HDL (mg/dL) 7.7 ± 26.1 [−2.16 to 17.7] 0.69 ± 23.5 [−7.26 to 8.64] NS Homocysteine (μmol/L) −6.04 ± 9.04 [−9.48 to −2.59] −2.75 ± 10.02 [−6.14 to 0.64] NS Ferritin (ng/mL) 23 (−107, 216.45) [−26.67 to 168.58] −31.4 (−153.65, 124.35) [−194.55 to 38.5] NS Total iron-binding capacity (μg/dL) −15 (−276, 103.5) [−322.35 to −0.74] 6 (−177, 162) [−88.11 to 128.17] NS Hb (g/dL) −0.23 ± 2.04 [−1.01 to 0.53] −0.14 ± 2.29 [−0.9 to 0.62] NS Albumin (g/dL) 0.61 ± 1.14 [0.17–1.05] 0.4 ± 1.09 [−0.01 to 0.81] NS NS, not significant. a Continuous data with normal distribution are expressed as means ± SDs [95% confidence interval] and continuous data with skewed distribution as median (IQR) [95% confidence interval]. Table 3. Adverse events in patients Adverse events Serious adverse events Other adverse events Selenium group a Placebo group Death due to myocardial infarction 2 2 Death due to cerebrovascular accident 1 0 Dyspepsia and abdominal pain 2 3 Nausea and vomiting 2 0 a Number of patients with adverse events. The beneficial effect of selenium supplementation on the nutritional status of patients with chronic diseases was also observed in a randomized interventional study by Federico et al. [39]; in this interventional study, aggravation of the nutritional status was prevented in patients with cancer of the digestive tract who received selenium and zinc supplementation compared with those who received placebo. 721 Selenium for malnutrition in hemodialysis patients ORIGINAL ARTICLES Selenium group Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 Measured parameters AC K N O W L E D G E M E N T S This study was funded by a Grant from Shiraz University of Medical Sciences, Shiraz, Iran. We would like to thank Dr Nasrin Shokrpour for English review of this article. C O N F L I C T O F I N T E R E S T S TAT E M E N T None of the authors has any conflict of interest to declare. (See related article by Thompson and Tonelli. Selenium for malnutrition in hemodialysis patients: have we considered all of the elements? Nephrol Dial Transplant 2013; 28: 498–500.) REFERENCES 1. Kalantar-Zadeh K, Ikizler TA, Block G et al. Malnutrition– inflammation complex syndrome in dialysis patients: causes and consequences. Am J Kidney Dis 2003; 42: 864–881. 2. Kalantar-Zadeh K, Stenvinkel P et al. Inflammation and nutrition in renal insufficiency. Adv Ren Replace Ther 2003; 10: 155–169. 3. Kalantar-Zadeh K. Recent advances in understanding the malnutrition–inflammation-cachexia syndrome in chronic kidney disease patients: what is next? Semin Dial 2005; 18: 365–369. 4. Himmelfarb J, Stenvinkel P, Ikizler TA et al. The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int 2002; 62: 1524–1538. 5. Locatelli F, Canaud B, Eckardt KU et al. Oxidative stress in endstage renal disease: an emerging threat to patient outcome. Nephrol Dial Transplant 2003; 18: 1272–1280. 6. Coombes JS, Fassett RG. Antioxidant therapy in hemodialysis patients: a systematic review. Kidney Int 2012; 81: 233–246. 7. Kuhlmann MK, Levin NW. Potential interplay between nutrition and inflammation in dialysis patients. Contrib Nephrol 2008; 161: 76–82. 8. Prabhu KS, Zamamiri-Davis F, Stewart JB et al. Selenium deficiency increases the expression of inducible nitric oxide synthase in RAW 264.7 macrophages: role of nuclear factorkappaB in up-regulation. Biochem J 2002; 366: 203–209. 9. Yavuz O, Bicik Z, Cinar Y et al. The effect of different dialysis membranes on oxidative stress and selenium status. Clin Chim Acta 2004; 346: 153–160. 10. Bonomini M, Forster S, De Risio F et al. Effects of selenium supplementation on immune parameters in chronic uraemic patients on haemodialysis. Nephrol Dial Transplant 1995; 10: 1654–1661. 11. Fujishima Y, Ohsawa M, Itai K et al. Serum selenium levels are inversely associated with death risk among hemodialysis patients. Nephrol Dial Transplant 2011; 26: 3331–3338. 12. Rucker D, Thadhani R, Tonelli M. Trace element status in hemodialysis patients. Semin Dial 2010; 23: 389–395. 13. Tonelli M, Wiebe N, Hemmelgarn B et al. Trace elements in hemodialysis patients: a systematic review and meta-analysis. BMC Med 2009; 7: 25. 722 M. Salehi et al. Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 ORIGINAL ARTICLES CVD is the main cause of mortality in HD population [4, 6]. Malnutrition, inflammation and oxidative stress are all proposed to contribute to the pathogenesis of CVD in HD patients through mediation of vascular injury and endothelial dysfunction [4, 6, 40]. Surrogate markers of malnutrition and inflammation such as MIS, SGA score and serum levels of IL-6 have been demonstrated to be correlated with the prospective mortality rate in HD patients in several studies [21, 22, 41, 42]. Moreover, antioxidant therapies like vitamin E and N-acetylcysteine have been shown to be effective in reducing CVD events in HD patients [43, 44]. Therefore, selenium supplementation that has been demonstrated to be effective in hampering the surrogate markers of malnutrition, inflammation and oxidative stress in our study could potentially reduce CVD and its resultant mortality in HD patients. However, this claim needs to be further evaluated in future studies with larger sample sizes and longer durations. Selenium has a low therapeutic index, and toxicity may occur with its high-dose supplementation; adverse effects of selenium toxicity include brittle hair and nails and their loss, dermatitis, pulmonary edema, neurotoxicity, nausea and vomiting [12, 45]. Except for gastrointestinal disturbances in few patients, none of the other adverse effects of selenium toxicity was observed in this trial. A few cardiovascular deaths also occurred in both selenium and placebo groups; however, these deaths are probably due to higher prevalence of CVDs in HD population. The low incidence of adverse events in this study may be due to the administration of lower dose of selenium (200 µg/day) compared with its maximum allowance dose (400–450 µg/ day) [12]. Other trials which used similar doses of selenium in HD patients also did not report the adverse events of selenium toxicity [18, 30, 32]. However, due to short duration and low sample size of these trials as well as our trial, selenium supplementation in HD patients could not be claimed safe. The main limitation of our study, and interpretation of its results, is the lack of the selenium measurement at the baseline and at the end of the treatment phase. Without measuring these levels, it is not known to what extent the differences in the baseline selenium levels might have impacted the therapeutic effect of the selenium supplementation; therefore, it is impossible to say that the selenium supplementation was actually successful. Low sample size and short duration are other limitations of our study. In conclusion, our study for the first time demonstrated the efficacy of selenium supplementation in improving the nutritional status of HD patients in short term. This perceived effect may be mediated by hampering oxidative stress and inflammation. Therefore, selenium supplementation may be considered as an addition to the current therapeutic remedies for malnutrition in HD patients. However, further multicenter trials with larger sample sizes are needed to confirm its efficacy and safety in the long term and also to delineate its possible mechanisms of action. 723 Selenium for malnutrition in hemodialysis patients ORIGINAL ARTICLES Received for publication: 29.1.2012; Accepted in revised form: 6.4.2012 Downloaded from https://academic.oup.com/ndt/article/28/3/716/1812238 by guest on 05 November 2022 30. Adamowicz A, Trafikowska U, Trafikowska A et al. Effect of erythropoietin therapy and selenium supplementation on selected antioxidant parameters in blood of uremic patients on longterm hemodialysis. Med Sci Monit 2002; 8: CR202–CR205. 31. Koenig JS, Fischer M, Bulant E et al. Antioxidant status in patients on chronic hemodialysis therapy: impact of parenteral selenium supplementation. Wien Klin Wochenschr 1997; 109: 13–19. 32. Ardalan MR, Tubbs RS, Shoja MM. Vitamin E and selenium co-supplementation attenuates oxidative stress in haemodialysis patients receiving intra-dialysis iron infusion. Nephrol Dial Transplant 2007; 22: 973–975. 33. Duntas LH. Selenium and inflammation: underlying antiinflammatory mechanisms. Horm Metab Res 2009; 41: 443–447. 34. Pei Z, Li H, Guo Y et al. Sodium selenite inhibits the expression of VEGF, TGFbeta(1) and IL-6 induced by LPS in human PC3 cells via TLR4-NF-(K)B signaling blockage. Int Immunopharmacol 2010; 10: 50–56. 35. Agbor GA, Vinson JA, Patel S et al. Effect of selenium- and glutathione-enriched yeast supplementation on a combined atherosclerosis and diabetes hamster model. J Agric Food Chem 2007; 55: 8731–8736. 36. Bunk MJ, Combs GF, Jr. Effect of selenium on appetite in the selenium-deficient chick. J Nutr 1980; 110: 743–749. 37. Ewan RC. Effect of selenium on rat growth, growth hormone and diet utilization. J Nutr 1976; 106: 702–709. 38. Olsson U. Impaired ketone body metabolism in the selenium deficient rat. Possible implications. Metabolism 1985; 34: 993–998. 39. Federico A, Iodice P, Federico P et al. Effects of selenium and zinc supplementation on nutritional status in patients with cancer of digestive tract. Eur J Clin Nutr 2001; 55: 293–297. 40. Kaysen GA. Association between inflammation and malnutrition as risk factors of cardiovascular disease. Blood Purif 2006; 24: 51–55. 41. Chan M, Kelly J, Batterham M et al. Malnutrition (subjective global assessment) scores and serum albumin levels, but not body mass index values, at initiation of dialysis are independent predictors of mortality: a 10-year clinical cohort study. J Ren Nutr 2012 (Epub ahead of print). 42. Honda H, Qureshi AR, Heimburger O et al. Serum albumin, Creactive protein, interleukin 6, and fetuin a as predictors of malnutrition, cardiovascular disease, and mortality in patients with ESRD. Am J Kidney Dis 2006; 47: 139–148. 43. Boaz M, Smetana S, Weinstein T et al. Secondary prevention with antioxidants of cardiovascular disease in endstage renal disease (SPACE): randomised placebo-controlled trial. Lancet 2000; 356: 1213–1218. 44. Tepel M, van der Giet M, Statz M et al. The antioxidant acetylcysteine reduces cardiovascular events in patients with end-stage renal failure: a randomized, controlled trial. Circulation 2003; 107: 992–995. 45. Fairweather-Tait SJ, Bao Y, Broadley MR et al. Selenium in human health and disease. Antioxid Redox Signal 2011; 14: 1337–1383. 14. Dworkin B, Weseley S, Rosenthal WS et al. Diminished blood selenium levels in renal failure patients on dialysis: correlations with nutritional status. Am J Med Sci 1987; 293: 6–12. 15. Liu ML, Xu G, Huang ZY et al. Euthyroid sick syndrome and nutritional status are correlated with hyposelenemia in hemodialysis patients. Int J Artif Organs 2011; 34: 577–583. 16. Malgorzewicz S, Rutkowski P, Jankowska M et al. Effects of renal-specific oral supplementation in malnourished hemodialysis patients. J Ren Nutr 2011; 21: 347–353. 17. Saghaei M. Random allocation software for parallel group randomized trials. BMC Med Res Methodol 2004; 4: 26. 18. Zachara BA, Gromadzinska J, Palus J et al. The effect of selenium supplementation in the prevention of DNA damage in white blood cells of hemodialyzed patients: a pilot study. Biol Trace Elem Res 2011; 142: 274–283. 19. Steiber AL, Kalantar-Zadeh K, Secker D et al. Subjective Global Assessment in chronic kidney disease: a review. J Ren Nutr 2004; 14: 191–200. 20. Steiber A, Leon JB, Secker D et al. Multicenter study of the validity and reliability of subjective global assessment in the hemodialysis population. J Ren Nutr 2007; 17: 336–342. 21. Kalantar-Zadeh K, Kopple JD, Block G et al. A malnutrition– inflammation score is correlated with morbidity and mortality in maintenance hemodialysis patients. Am J Kidney Dis 2001; 38: 1251–1263. 22. Rambod M, Bross R, Zitterkoph J et al. Association of malnutrition–inflammation score with quality of life and mortality in hemodialysis patients: a 5-year prospective cohort study. Am J Kidney Dis 2009; 53: 298–309. 23. Nursal TZ, Noyan T, Tarim A et al. A new weighted scoring system for Subjective Global Assessment. Nutrition 2005; 21: 666–671. 24. Zal F, Mostafavi-Pour Z, Vessal M. Comparison of the effects of vitamin E and/or quercetin in attenuating chronic cyclosporine A-induced nephrotoxicity in male rats. Clin Exp Pharmacol Physiol 2007; 34: 720–724. 25. Del Vecchio L, Locatelli F, Carini M. What we know about oxidative stress in patients with chronic kidney disease on dialysis —clinical effects, potential treatment, and prevention. Semin Dial 2011; 24: 56–64. 26. Zachara BA, Gromadzinska J, Wasowicz W et al. Red blood cell and plasma glutathione peroxidase activities and selenium concentration in patients with chronic kidney disease: a review. Acta Biochim Pol 2006; 53: 663–677. 27. Guo CH, Wang CL, Chen PC et al. 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. 28. Saint-Georges MD, Bonnefont DJ, Bourely BA et al. Correction of selenium deficiency in hemodialyzed patients. Kidney Int Suppl 1989; 27: S274–S277. 29. Richard MJ, Ducros V, Foret M et al. Reversal of selenium and zinc deficiencies in chronic hemodialysis patients by intravenous sodium selenite and zinc gluconate supplementation. Timecourse of glutathione peroxidase repletion and lipid peroxidation decrease. Biol Trace Elem Res 1993; 39: 149–159.