Next Article in Journal
Oxidative Status, Iron Plasma Levels in Venous Thrombosis Patients
Previous Article in Journal
Role of Uremic Toxins, Oxidative Stress, and Renal Fibrosis in Chronic Kidney Disease
Previous Article in Special Issue
Induction of Neuroinflammation and Brain Oxidative Stress by Brain-Derived Extracellular Vesicles from Hypertensive Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Selenium as a Modulator of Redox Reactions in the Prevention and Treatment of Cardiovascular Diseases

1
Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
2
Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(6), 688; https://doi.org/10.3390/antiox13060688
Submission received: 29 March 2024 / Revised: 21 May 2024 / Accepted: 23 May 2024 / Published: 3 June 2024
(This article belongs to the Special Issue Antioxidant Therapy for Cardiovascular Diseases)

Abstract

:
Cardiovascular diseases stand as the predominant global cause of mortality, exerting a profound impact on both life expectancy and its quality. Given their immense public health burden, extensive efforts have been dedicated to comprehending the underlying mechanisms and developing strategies for prevention and treatment. Selenium, a crucial participant in redox reactions, emerges as a notable factor in maintaining myocardial cell homeostasis and influencing the progression of cardiovascular disorders. Some disorders, such as Keshan disease, are directly linked with its environmental deficiency. Nevertheless, the precise extent of its impact on the cardiovascular system remains unclear, marked by contradictory findings in the existing literature. High selenium levels have been associated with an increased risk of developing hypertension, while lower concentrations have been linked to heart failure and atrial fibrillation. Although some trials have shown its potential effectiveness in specific groups of patients, large cohort supplementation attempts have generally yielded unsatisfactory outcomes. Consequently, there persists a significant need for further research aimed at delineating specific patient cohorts and groups of diseases that would benefit from selenium supplementation.

1. Introduction

Cardiovascular disease (CVD) is the primary cause of global mortality and significantly contributes to a decline in quality of life [1,2,3]. Based on statistical data, in 2019, the mortality attributed to CVD was 17.8 million, and projections suggest that by 2030, this figure is expected to increase to 23 million [2,3]. In order to reduce the prevalence of CVD within the population, it is important to take into consideration not only the spectrum of CVD risk factors but also the underlying pathomechanisms governing the progression of these disorders [4]. Increased oxidative stress is recognized as an important contributor to CVD pathogenesis, precipitating conditions including myocardial infarction, ischemia or reperfusion injury, and heart failure [5]. The excessive generation of reactive oxygen species (ROS) leads to a reduction in nitric oxide bioavailability, thereby promoting vasoconstriction and eliciting arterial hypertension. Additionally, ROS disrupt calcium homeostasis in the myocardium, resulting in arrhythmias and facilitating cardiac remodeling through signaling hypertrophy and apoptosis. Moreover, ROS are key factors in instigating the formation of atherosclerotic plaques [4].
In recent years, numerous studies have explored the use of antioxidants as therapeutic agents. One particular trace element, Se, has garnered special attention [6,7]. Se holds a significant and unique role among microelements. It serves as an essential component of the 21st amino acid, selenocysteine (SeC), which is found within the catalytic sites of selenium-dependent enzymes. Proteins containing at least one SeC residue are termed selenoproteins and play numerous crucial physiological roles, primarily centered on maintaining cellular redox balance [8].
In addition to its antioxidant properties, Se can affect cell survival through the expression of proapoptotic proteins and regulation of autophagy in myocardial cells. Activation of caspase family genes leads to apoptosis both through the death receptor and through the mitochondrial pathway, in which the antiapoptotic factor BCL-2 is a major regulator [9]. Se deficiency leads to an increase in mRNA levels of caspase-3 and caspase-9 and a decrease in BCL-2 levels, which promotes cardiomyocyte death [10]. In addition, Se deficiency results in reduced expression of potassium channels, STAT3 activity, and mitochondrial function [11]. The STAT/JAK pathway mediates inflammation-related cytokines, regulating growth, survival, and differentiation. Se supplementation by increasing STAT3 protein may play a cardioprotective role after ischemia-reperfusion injury [12,13].
A comprehensive collection of 25 selenoprotein genes has been documented, which are distributed throughout various organs and tissues displaying diverse substrate specificity [14]. Among these proteins, there are five glutathione peroxidases, three iodothyronine deiodinases, three thioredoxin reductases, selenophosphate synthetase 2, methionine sulfoxide reductase B1, and selenoproteins F, H, I, K, M, N, O, P, S, T, V, and W [15,16]. The diverse occurrence of selenoproteins throughout the body enables them to potentially modulate pathogenetic mechanisms in numerous disease processes (Table 1, Figure 1) [14]. The significance of heart-specific selenoproteins is underscored by Benstoem et al., who demonstrated that low Se levels and subsequent removal of selenoproteins through deletion of SeC tRNA in the heart and skeletal muscles resulted in sudden cardiac arrest due to increased oxidative stress and inflammation [17].
Despite recent advances in CVD therapies, there are still many factors that need to be considered. The effect of dietary supplementation is an understudied topic; however, research suggests that it may influence the progression of CVD. This review illustrates the antioxidant role of Se, shedding light on its influence on the pathomechanisms underlying arterial hypertension, coronary heart disease, heart failure, and arrhythmias.
Table 1. Types of selenoproteins in humans and their functions and role in various diseases.
Table 1. Types of selenoproteins in humans and their functions and role in various diseases.
Proteins with SeFunctionsRelated DisordersReferences
Selenoproteins
FInvolvement in ER redox protein folding processColorectal cancer
Gastric cancer
[18]
Regulation of ER stress
HRegulation of redox homeostasisGastrointestinal tumor[19,20]
Protection against UVB-induced apoptosis
Protection against oxidative stress
Inhibition of cellular aging
Maintenance of genome integrity
Suppression of oxidative. DNA damage in tumorigenesis
IInvolvement in membrane lipid synthesis of phosphatidylethanolamine (PE) and plasmenyl PEHereditary spastic paraplegia[21,22]
Neuronal development and myelin synthesis
Regulation of T-cell function
KDegradation of misfolded proteinsMelanoma
Gastric cancer
Alzheimer’s disease
Atherosclerosis
[21,23,24,25]
Regulation of redox equilibrium
Antioxidant activity in cardiomyocytes
Modulation of calcium homeostasis in immune cells
Regulation of neuronal apoptosis induced by ER stress
Palmitoylation
MAntioxidantAlzheimer’s disease
Non-alcoholic fatty liver Disease
Solid tumors
[26,27]
Hypothalamic leptin signaling
Bone development
Neuroprotective
Role in body weight and energy metabolism
Calcium homeostasis
NOxidative and calcium homeostasis in endoplasmic reticulumSEPN1-related myopathies[28,29]
Muscle regeneration and satellite cell maintenance in skeletal muscle
OProper cellular response to oxidative stress
Transfers AMP from ATP to Ser, Thr, and Tyr residues on protein substrates (AMPylation)
Staphyloenterotoxemia
Ritter’s disease
[30]
PAntioxidantCancers
Ataxia and seizures
CVDs
[31,32,33]
Maintain neuronal activity
Regulate pancreatic β cell function
Transport Se to tissues
RAntioxidantCataracts[34]
Repairing oxidized proteins
SRegulate inflammationHashimoto’s
Thyroiditis
CVDs
[35,36]
Induce ER stress apoptosis
Immune regulation
TAntioxidant role in cardiomyocytesParkinson’s disease
Embryonic lethality in mice
[37,38]
Protection of the heart against ischemia/reperfusion injury
Hormone synthesis
Calcium mobilization
Neuroprotective
Control of glucose homeostasis
VSpermatogenesisAzoospermia[15,39]
WOxidationOsteoporosis
Anemia
[40,41]
Regulate bone metabolism
Support erythroblast and muscle development
Enzymes
Iodothyronine deiodinases (DIO)Act as biocatalysts for the regulation of the activity
of thyroid hormones
[42]
DIO1Participation in the production of the active thyroid hormone T3Various thyroid-related disorders[14,42]
Conversion of T4 to a reduced form T3 (rT3)
DIO2Participation in the production of the active thyroid hormone T3 Various thyroid-related disorders [14]
DIO3Contributes to the generation of the inactive rT3 and T2Various thyroid-related disorders [14,42]
Conversion of T4 to rT3 via inner-ring deiodination of T4
Thioredoxin
reductases (TrxR)
Involved in cellular antioxidative defense systems
and the maintenance of intracellular redox states to
maintain cell viability
[14]
TrxR1Redox regulationCancers[43,44]
Antioxidant defense
Maintenance of intercellular reducing conditions
TrxR2Redox signalingBone metabolic diseases
Embryonic lethality in mice
[45,46]
Metabolism, proliferation, differentiation, migration, and apoptosis of cells
RGR (Retinal G Protein Coupled Receptor)Involved in the visual cycle-regeneration of visual pigments in the retinaRetinitis pigmentosa[47,48]
Receptor for all-trans- and 11-cis-retinal
Glutathione peroxidases (GPx)
GPx1AntioxidantAtherosclerosis
Myocardial infarction
Alzheimer’s disease
Parkinson’s disease
Cerebral ischemia
Breast cancer
Diabetes mellitus type 2
[49,50,51,52,53]
GPx2AntioxidantColorectal cancer
Breast cancer
Prostate cancer
Liver cancer
Non-small-cell lung cancer
[54,55]
Inflammation prevention
Mucosal homeostasis
GPx3AntioxidantColorectal cancer
Gastric cancer
Thyroid cancer
[56]
GPx4AntioxidantSystemic lupus erythematosus
Neurodegenerative diseases
[57,58,59]
Ferroptosis regulator
Spermatogenesis
GPx6AntioxidantUnknown[60]
Figure 1. Effects of selenium on human body [61].
Figure 1. Effects of selenium on human body [61].
Antioxidants 13 00688 g001

2. The Application of Se in the Prevention and Treatment of CVD

2.1. Hypertension

Hypertension stands as a significant global health concern, as it is a leading cause of mortality worldwide, accounting for around 14% of all deaths [62]. The World Health Organization (WHO) estimates that hypertension affects over one billion people globally, with its prevalence steadily increasing over the past few decades [63].
Characterized by persistently elevated blood pressure levels, hypertension is a major risk factor for various adverse health outcomes, including heart disease, stroke, and kidney failure. Dietary factors, including high sodium intake, low potassium intake, excessive alcohol consumption, and diets high in saturated fats and processed foods, have been identified as significant contributors to the development and progression of hypertension [64]. Additionally, trace elements have garnered increasing attention in recent studies as potential contributors to hypertension. Selenoproteins, containing SeC as a prominent Se compound, act as antioxidants and regulators of redox processes, protecting cells from oxidative stress and free radical damage [65]. Immune system functionality, oxidative stress, and inflammatory reactions are involved in the onset and advancement of hypertension [66].
The molecular mechanisms underlying hypertension remain unclear, although inflammation has been implicated in its onset and progression [67]. Studies have shown a significant association between inflammatory markers and hypertension risk, with elevated levels of hs-CRP and IL-6 linked to increased disease incidence [68]. Se supplementation has been found to reduce CRP and IL-6 plasma concentrations by controlling the expression of selenoprotein genes and preventing NF-κB activation [69]. Additionally, Se may enhance nitrogen oxide (NO) synthesis by stimulating NO synthase activity [70] and inhibiting NO degradation, thereby promoting vasodilation [70,71]. Arterial stiffness, which also plays a significant role in hypertension, is associated with vascular remodeling characterized by changes in vessel structure and function [72]. Inflammatory factors, including IL-1, IL-17, and IL-6, are stimulated by Angiotensin II signaling, further contributing to vascular remodeling and arterial stiffness [73]. Anti-inflammatory properties of Se may help to mitigate vascular remodeling, reduce inflammation, and lower the risk of hypertension.
In a study conducted by Bastola et al. in 2020, analyzing data from 6683 participants in the National Health and Nutrition Examination Survey (NHANES) spanning from 2011 to 2016, it was observed that there exists a significant positive correlation between serum Se levels and hypertension, regardless of age or intake of antihypertensive medications [74]. The analysis revealed that serum Se levels at or above 120 μg/L (with a reference range about 70–150 μg/L) were notably associated with hypertension (OR = 1.46, 95% CI = 1.29–1.66) after adjusting for potential confounding variables [74]. Moreover, an elevated serum Se level exceeding 150 μg/L exhibited a strengthened association with hypertension (OR = 1.69, 95% CI = 1.32–2.17) [74]. Furthermore, the results indicated that an increase in serum Se levels was more pronounced in diastolic blood pressure measurements compared to systolic blood pressure measurements [74]. This observation aligns with previous findings reported by Mark et al., where dietary Se supplementation in a nutritionally deficient population resulted in the development of diastolic hypertension without affecting systolic blood pressure [75].
However, the influence of serum Se levels on hypertension risk remains a topic of controversy. Recent research contradicts the findings of Bastola et al. [74], suggesting that elevated Se consumption is associated with a decreased risk of hypertension.
Wu et al.’s research investigates the relationship between dietary Se intake and hypertension, marking a novel addition to previous studies that mainly focused on serum Se levels. Data from NHANES spanning from 2003 to 2018 were utilized to examine the link between dietary Se intake and the occurrence of hypertension in adults. Among 32,928 participants with an average dietary Se intake of 1.12 ± 0.53 μg, the overall hypertension prevalence was 36.55% [76]. Hypertension rates decreased across higher quartiles of dietary Se intake. Each quartile increase in dietary Se intake was associated with an 11% reduced likelihood of hypertension prevalence (OR = 0.89; 95% CI: 0.80–1.00; p = 0.0425) [76]. Subgroup analyses indicated no significant correlations between gender, age, body mass index, smoking status, alcohol consumption, diabetes mellitus, and the association between dietary Se intake and hypertension prevalence [76].
As these investigations were epidemiological in nature and relied on self-reported dietary data from surveys, susceptible to subjective bias, they established an association between serum Se levels or Se intake and hypertension, without necessarily establishing a direct cause-and-effect relationship. In contrast, a recent experimental study revealed that rats subjected to a low-selenium diet developed hypertension due to decreased urine sodium excretion [77]. The hypertension observed in Se-deficient rats was linked to elevated renal angiotensin II type 1 receptor (AT1R) expression and activity, which contributed to increased reabsorption of sodium in renal tubules [77]. The decreased Se intake resulted in reduced levels of GPx1, a selenoprotein, which via the NF-κB pathway leads to the up-regulation of AT1R expression [77].
In 2021, a study aimed to investigate the relationship between serum Se levels and mortality in hypertensive patients [78]. Utilizing data from NHANES 2003–2004, participant outcomes were tracked until December 31, 2015. A total of 929 individuals were included in the analysis, with 307 deaths observed, including 56 cardiovascular fatalities, over an average follow-up period of 121.05 ± 40.85 months. A U-shaped association was noted between serum Se levels and both all-cause and cardiovascular mortality. Participants were stratified into quartiles based on serum Se levels (Q1: ≤124 μg/L, Q2: 125–135 μg/L, Q3: 136–147 μg/L, Q4: ≥148 μg/L) [78]. In a fully adjusted model, comparisons among quartiles revealed that individuals in the third quartile (Q3) had lower risks of both all-cause [HR (95% CI), 0.57 (0.39–0.81)] and cardiovascular mortality [HR (95% CI), 0.33 (0.13–0.86)] [78]. The lowest mortality rates for all-cause and cardiovascular deaths were observed at serum Se levels of 136 μg/L and 130 μg/L, respectively [78].
Se therapy holds promise as a potential agent in managing hypertension, a prevalent global health issue associated with significant morbidity and mortality. While epidemiological studies suggest a correlation between serum Se levels and hypertension risk, experimental research in animal models has provided physiological insights into the role of Se in blood pressure regulation. However, the exact nature of the relationship between Se and hypertension requires further investigation, including well-designed clinical trials to assess the efficacy and safety of Se supplementation in hypertensive individuals. A better understanding of selenium’s therapeutic potential in hypertension may offer new strategies for prevention and treatment in clinical practice.

2.2. Coronary Heart Disease

Coronary heart disease (CHD) is a common CVD that is one of the leading causes of morbidity and mortality worldwide. Complex interactions between multiple genetic and environmental factors are involved in its pathogenesis [79]. An important factor in the development and progression of CHD is oxidative stress, which contributes to endothelial dysfunction. Consequently, antioxidants, especially those from the diet, have become the focus of several studies for their role in the prevention of CHD.
Se is a trace element that, through the activity of GPx and other selective selenoproteins, prevents lipid peroxidation and oxidative stress and presents anti-inflammatory effects [80]. It has a significant role in both the acute and chronic phases of CHD. Se plays a crucial role in the neutralization of reactive oxygen and nitrogen species, contributing to the reduction in ischemic injury and left ventricular hypertrophy. In the literature, Se deficiency is found in most patients after myocardial infarction (MI) [81,82]. In a clinical study of 84 patients with acute MI, lower levels of Se were observed in plasma, erythrocytes, and nails compared to controls [83]. Furthermore, another study observed in patients with acute MI a significant correlation between baseline Se levels and peak troponin I levels, which is linked to myocardial necrosis, suggesting an association between Se changes and the extent of MI [84]. Venardos et al. revealed in a rat model that the hearts of selenium-deficient animals were more prone to ischemia-reperfusion injury with increased protein and lipid peroxidation [85]. On the other hand, Zachara et al. observed that blood Se levels in patients with MI did not differ compared to the control group and remained stable throughout the study period [86].
The effect of Se on the development of CHD and its association with mortality remains controversial. A meta-analysis by Flores-Mateo et al. revealed a statistically significant inverse association between Se levels and CHD risk in observational studies [87]. They revealed that a 50% increase in Se levels corresponded to a 24% reduction in coronary incident risk. Additionally, this meta-analysis showed that the pooled relative risk of CHD based on a ratio of the highest and lowest Se levels was 0.85 (95% CI, 0.74–0.99). However, the validity of the association in the observational studies analyzed was uncertain [87]. Moreover, Kuria et al. showed that physiologically high levels of Se in the body were related to reduced CHD mortality (RR¼ 0.73, 95% CI: 0.59–0.91), as well as reduced CHD incidence (RR¼ 0.83, 95% CI: 0.74–0.94) [88]. Higher plasma Se concentrations were correlated with lower CHD risk in a nested case–control study in prospective Dongfeng-Tongji cohort [89]. An evaluation of the non-linear relationship of dietary Se intake and serum Se concentration with CHD incidence was conducted by Xie et al. This study included data on 17,867 participants from the United States collected from NHANES between 1999 and 2006. It showed a negative correlation between serum Se concentration and CHD (OR: 0.989, 95% CI: 0.981, 0.997, p = 0.006), which was nonlinear after adjusting for multiple variables. Participants with higher serum Se levels had a reduced incidence of CHD, and with further increases in serum Se levels, the incidence of CHD decreased slowly. Furthermore, Xie et al. observed a U-shaped curve representing the relation between dietary Se intake and all-cause mortality. In contrast, there was no statistically significant association between dietary Se intake and the risk of developing CHD (p = 0.206) in an analysis including confounders [90].
In other studies, there was no significant correlation between decreased Se levels and increased CHD risk or mortality [91,92,93]. It has been suggested that discrepancies between studies may be due to differences in baseline Se levels in subjects depending on geographic region. The protective effects of Se are observed in regions with low Se levels, while in individuals with sufficient Se levels, supplementation may have no additional benefit. Other reasons for the inconsistent findings include socioeconomic factors, differences in patients’ risk profiles, and other variables affecting Se concentrations [88,93,94,95]. A meta-analysis of 16 randomized controlled trials involving 43,998 participants showed that Se supplementation did not reduce coronary heart disease mortality or affect lipid profile. Nevertheless, it revealed a substantial effect of increased Se supply on the increase in GPx activity (SMD = 0.5; 95% CI, 0.36–0.64; p < 0.001) and on the decrease in serum CRP levels (SMD = −0.48; 95% CI, −0.96 to 0; p = 0.049), which is one of the risk factors for CHD due to its direct correlation with the presence and severity of coronary atherosclerosis [96]. Similar results on the effect on GPx activity in endothelial cells were obtained by Schnabel et al. based on in vitro cell culture and in vivo clinical trial data [97].
Evidence suggesting an association between blood Se and CHD remains insufficient. Further studies in larger patient cohorts are needed to investigate selenium-dependent effects at the molecular level to provide reliable data on the preventive and diagnostic use of Se in CHD in clinical practice.

2.3. Heart Failure

Heart failure (HF) stands as a significant global health concern, marked by persistent challenges in enhancing patient quality of life and long-term survival despite advancements in therapeutic interventions. HF is defined as a functional and structural abnormality of the cardio-vascular system, which results in increased pressure in the heart or a deficient cardiac output during resting or exercising [98]. Within the pursuit of novel strategies for HF prevention and management, attention has turned towards the potential impact of Se.
Se emerges as a pivotal element in both the prevention and treatment paradigms of HF, yet its precise role warrants comprehensive examination. Understanding the significance of Se and its correlation with HF is essential. Fundamental to note are the diverse pathophysiological mechanisms, including oxidative stress, inflammation, and nutrient deficiencies, all contributing to the development and progression of HF.
There are many publications dedicated to the topic of Se deficiency and HF. The study of Mirdamadi et al. showed an association between low level of Se and a high incidence of HF [99]. A total of 32 hospitalized patients with HF and 32 healthy controls were enrolled in the case–control study. The serum Se levels were compared in case and control groups. The mean serum Se was 92.5 ± 22.44 mg/dL in patients with HF and 109.3 ± 29.62 mg/dL in healthy controls (p = 0.013). This study showed statistically significant lower levels of serum Se in patients with HF in comparison to healthy individuals. Furthermore, a noteworthy negative correlation was noted between Se levels and both left ventricular volume and pulmonary artery pressure (rho = −0.39, p= 0.03 and rho = −0.45, p = 0.01, respectively). Additionally, although not statistically significant, a positive trend was noted between the Se level and left ventricular ejection fraction. This observation potentially indicates the impact of Se deficiency on cardiac function and HF pathogenesis [99].
A prospective cohort study by Bomer elucidates the adverse implications of Se deficiency on patient outcomes, with impaired mitochondrial function and oxidative stress underscored as potential underlying mechanisms. The study enrolled 2516 patients with worsening HF to investigate Se deficiency in their serum. Additionally, the investigation dived into potential mechanisms by which Se deficiency might affect prognosis, by culturing human cardiomyocytes without Se and assessing mitochondrial function and oxidative stress. In cultured human cardiomyocytes, Se deprivation impaired mitochondrial function and oxidative phosphorylation, which led to an increase in intracellular ROS levels [100].
The study revealed that over 20% of patients had serum Se concentration under <70 μg/L. Moreover, the Se deficiency was associated with poorer quality of life, exercise capacity, and worse prognosis. Serum concentrations of 70–100 μg/L had similar adverse associations. Notably, supplementation trials exhibit promising amelioration of clinical symptoms in HF patients, hinting at selenium’s therapeutic potential [100]. Recent meta-analysis performed by Jenkins, which used data from 43 different studies (N = 114,146), examined the effect of antioxidant mixtures in relation to mortality. Antioxidant mixtures were defined as a combination of two or more of the following molecules: vitamin A, retinol, β-carotene, vitamin C, vitamin E, Se, zinc, and copper. They concluded that there was a significant reduction in both cardiovascular and all-cause mortality when Se was included in the antioxidant mixture (RR 0.77; 95% CI 0.62, 0.97 and RR 0.90; 95% CI 0.82, 0.98, respectively) [101].
These results were not observed when Se was not part of the antioxidant mixture [101]. However, the existing body of evidence remains insufficient to advocate widespread Se supplementation in HF management, necessitating further high-level research to delineate its precise role and therapeutic efficacy. Prospective cohort studies are warranted to ascertain the causative link between Se deficiency and HF incidence, shedding light on the nuanced interplay between Se and cardiac homeostasis.
In conclusion, Se emerges as a promising adjunct in HF management, holding potential for enhancing patient outcomes. While its precise mechanisms and therapeutic implications necessitate further elucidation, Se supplementation holds promise as a viable strategy for HF intervention, pending rigorous validation through high-level evidence. Efforts to unravel the intricate interplay between Se deficiency and HF pathophysiology remain imperative, paving the way for optimized therapeutic strategies and improved patient care.

2.4. Arrhythmias

Atrial fibrillation (AF) is the most common cardiac arrhythmia. Its prevalence varies from 2% to 12%, gradually increasing over age [102]. Numerous contributing factors have been postulated to intricately participate in the pathomechanism of AF, including inflammatory processes, fibrotic remodeling, and heightened oxidative stress [103]. Excessive ROS generation is considered a direct factor affecting propagation of action potential and functioning of the ion channels [104]. As a result of these findings, attempts were made to determine the role of microelements such as Se in AF development and the potential impact of its supplementation in antiarrhythmic treatment.
In the PREVEND study, a total cohort of 6000 patients with urinary albumin excretion ≥10 mg/L and 2592 participants with excretion less than 10 mg/L were qualified for evaluation in 3-year intervals [105]. On each control visit, a venous blood sample was collected, and a patient underwent ECG examination. A total of 6894 patients turned up on a second visit, of which 5452 had available measurements of micronutrients concentration including Se [106]. A mean follow-up was 6.2 years, during which 136 participants developed AF. Of 3900 non-smoking participants with Se deficiency established as <70 μg/L, the risk of developing AF was 68% higher than in the general population (95% CI 1.09 to 2.59, p = 0.018). In a group above the age of 60, this association remained significant also among smoking participants. Lowered Se levels were also encountered in patients with newly diagnosed AF compared to healthy subjects in study by Ardahanli and Ozkan [107]. However, this association was statistically significant only in men but not in women. This was attributed to differences in nutrition and concurring risk factors. Bazargani et al. observed that patients with AF have not only lowered the level of Se but also zinc, while the level of copper decreased [108].
Measurements of Se were also conducted during episodes of paroxysmal AF and were evaluated 24 h and 28 days after restoration of sinus rhythm [109]. Statistically significant lowered values were obtained only during arrhythmic episodes, remaining unaltered at further evaluations. All these findings are not surprising given the fact that fibrosis, inflammatory mediators, and oxidative stress are predisposing factors to arrhythmias. Each 10% increase in redox potential of glutathione is associated with a 40% risk of an AF incident [110].
There is still a lack of proper studies covering isolated use of Se in treatment of AF. Mirhoseini et al. analyzed data from 4699 critically ill patients admitted to the intensive care unit [111]. The group, which received a mix of antioxidants (i.e., vitamin C, vitamin E and Se), did not display better outcomes in terms of incidence reduction in atrial arrhythmias compared to controls in the first two weeks of hospitalization (3.02% vs. 3.31%, p = 0.62). However, the antioxidant group also did not display decreased mortality; it had longer expected survival time. Vitamins C and E were shown to reduce risk of AF recurrence after successful cardioversion but also to treat ventricular arrhythmias and arrhythmias linked with long-QT interval. Therefore, further trials are necessary in order to pinpoint effects of Se use in arrhythmic diseases besides AF [112].
In addition to its idiopathic etiology, AF is frequently recognized as a complication after both cardiac, thoracic, and to a lesser extent, other types of surgeries. Most commonly, it occurs within 48 h after the procedure and typically is a transient phenomenon. Its incidence was estimated from 10% up to 60% of cardiac surgeries, depending on the type of procedure with the highest risk related to valve replacement. The risk progressively increases in line with age [113]. Its pathomechanism is not yet completely understood. It is known that both acute surgical factors and progressive remodeling contribute to its development. Excessive oxidative stress and altered mechanisms of its prevention are considered to be two of the major factors [114]. Among patients who underwent intermediate risk coronary artery surgery, it was shown that low preoperative Se is associated with postoperative AF [72]. On the other hand, Kyaruzi et al. found no statistically significant difference in concentration level of Se between patients with and without AF [115].

2.5. Cardiac Surgeries

Se is recognized as a factor affecting outcomes of cardiac surgeries. Koszta et al. observed that patients who died following cardiac surgery had lower levels of blood Se compared to survivors (mean 102.2 μg/L vs. 111.1 μg/L, p = 0.047) [116]. It was considered a minor risk factor of postoperative mortality. It is yet unclear whether Se supplementation would be beneficial in terms of survival.
In a prospective study by Stoppe et al., sixty patients were evaluated on concentration of Se, copper, and zinc before and after cardiac surgeries with the use of a cardiopulmonary bypass [117]. Se concentration measured one hour after admission to the intensive care unit showed that it is an independent predictor of postoperative multiorgan failure occurrence within the next 48–72 h. Among patients with no organ failure, its level was 72.2 μg/L on average, while in patients with multiorgan failure, it was 61.5 μg/L. Se concentration also correlated with length of stay and perioperative inflammation. A similar pattern was also reported by Zhou et al., but there was no difference in concentration of Se between groups with AF and those who were in sinus rhythm [118]. The levels of Se after open-heart surgery also varied with regard to duration of a procedure, length of intensive care unit stay, and duration of cardiopulmonary bypass [119]. Similarly, Stevanovic et al. found low levels of Se as a predictive factor for developing complications and negative correlation to myocardial damage monitored by levels of CK-MB (r = −0.571, p < 0.001) [120].
There were attempts to determine whether Se perioperative administration would be beneficial in reducing post-surgical adverse effects. However, no significant differences were observed in the reduction in mortality, acute kidney injury, length of hospitalization and intensive care unit stay, CK-MB, and troponin I levels compared to placebo groups [121]. SUSTAIN CSX trial assessed the ability of selenite treatment in reducing postoperative organ dysfunction and mortality in 1416 cardiac surgery patients. Their study did not confirm the benefits of perioperative Se supplementation in patients with high cardiac risk [122]. However, Schmidt et al. managed to obtain a reduction in postoperative vasoactive support with a high dose of sodium selenite [123]. The impact of Se supplementation on the metabolic status of patients undergoing coronary artery bypass grafting was examined by Kamali et al. [124]. A four-week intake of Se at a dose of 200 μg/day resulted in an elevation of HDL cholesterol levels and glutathione and a reduction in fasting plasma glucose, insulin, malondialdehyde, and high sensitivity CRP [124].
Although there are available data from clinical trials assessing the effectiveness of single Se supplementation, its usage in combination with other agents still requires further evaluation [121].

3. The Influence of Selenium in the Environment and Selenium-Enriched Diet on Human Health

Adequate levels of Se ensure homeostasis not only in cardiovascular systems, but also endocrine, reproductive, and nervous systems [125,126]. In food, Se is typically present in trace amounts, measuring ≤ 1 mg/kg, with some exceptions such as Brazilian nuts, which can contain even 20 mg Se per kg [127]. The content of Se in various foods is presented in Figure 2. The recommended Se intake depends on age, sex, and physiological status. For adults, the recommended daily intake ranges between 55 and 70 µg/day [128,129]. Pregnant women should aim for 65 µg/day, while breastfeeding mothers require slightly more Se, approximately 75 µg/day [130]. In healthy individuals, the plasma Se concentrations around 100 µg/L recorded the highest selenoprotein activity [131]. It is essential what the maximum tolerable intake is, because of side effects that may occur [126].
Organic forms of Se are typically encountered as sulfur amino acid analogs, notably selenomethionine and SeC, while inorganic Se exists in the form of Se salts, selenate (SeO−24) and selenite (SeO−23) [134]. Both organic and inorganic forms can serve as valuable dietary sources; however, the inorganic forms appear to be more toxic than the organic ones [135]. The inorganic forms of Se are mostly found in seafood, legumes, and dry fruits [127]. They are mostly used in biofortification and in supplements. The selenate, emerging as the most prevalent one, is detected primarily in water [136], but can also be found in wheat straw [137]. The organic variants dominate in animal products, such as beef and poultry. Apart from that, selenomethionine can also be found in vegetables, grains, legumes, nuts, and yeast, along with being present in dietary supplements [138]. SeC is present in poultry, beef, and dairy products, while selenium-methylselenocysteine is primarily detected in Allium vegetables like garlic, onion, broccoli, and leeks [139,140]. The primary factor influencing Se concentration in plant-based foods, and to some extent in animal-derived ones, is the availability of Se in the environment, especially in the soil [141]. Se is present in both soil and water, where it can be mobilized and incorporated into the food chain through plant roots or aquatic organisms. This phenomenon raises concerns about potential long-term effects on human health [142,143].
Depending on the region of crop cultivation and animal husbandry, the Se content in foods may vary [144]. This variability necessitates careful consideration when devising strategies for Se supplementation. However, it is worth noting that the Se concentration in water and soil is not the sole factor influencing its uptake by plants. Soil Se mobility processes and other factors such as soil pH or organic matter content also significantly affect its availability to plants (Figure 3). Plants uptake Se in organic forms, such as selenate and selenite, to a much greater extent than in inorganic forms, and some plant species have the ability to accumulate this microelement in high concentrations, making them Se hyperaccumulators [145]. The environmental role of Se in the atmosphere and its effect on human health is an understudied topic. Studies suggest that in general, atmospheric background levels of Se are low in urban areas [145]. Further research is necessary to assess in greater detail Se levels in rural areas and its role in agriculture. This is associated with selenium’s potentially toxic effects on natural ecosystems due to its bioaccumulation potential. Se found in water and soil can be mobilized to enter the food chain, which is coupled with concerns about its long-term environmental effects on humans and ecosystems [145]. Most toxicity levels are based on absorption in the aquatic environment, which is much lower in natural populations than absorption in the food chain [146]. According to Tan et al., 40% of Se emissions in the atmosphere and aquatic environment come from human industrial activities such as mining, refineries, agriculture, and coal-fired power plants [147]. Overall, the limited existing research indicates that monitoring of environmental Se levels should be considered. This would further aid in developing appropriate management strategies to minimize the risk of toxicity to humans.
The pharmacokinetics of Se therapy and the general health impact depend not only on the chemical form of this trace element but also its bioavailability and the mode of administration [149]. It is important to underline that organic Se, particularly SeMet, is characterized by higher bioavailability [128,150] and offers more benefits than its inorganic counterpart within the context of oral supplementation and a well-rounded diet [151]. The most efficacious parenteral forms of Se supplementation are inorganic selenocompounds, and its organic forms often are not available for intravenous use [152].
Excessive Se intake leads to a change in the mechanism of action of this trace element on tissues; it becomes a pro-oxidant and, as a result, causes oxidative harm to cells and tissues and occurring side effects [126,153]. However, the toxic one-time dose is not precisely known [154]. The lethal one-time dose seems to oscillate around 4000 μg [154]. Acute toxicity manifests with diarrhea, fatigue, stomach and joint pain, respiratory issues, nausea, and hair loss [155].
Considering the chronic delivery of Se to the body, the lowest observed adverse effect levels of 3–4 µg Se/kg body weight/day in humans are close to normal consumption levels, ranging from 0.15 to 4 µg/kg body weight/day. Consequently, Se can be viewed as a trace element with a narrow concentration range in the human body, spanning from deficiency to optimal physiological levels and toxicity [149]. The chronic ingestion of excessive Se leads to selenosis. This condition manifests with a range of symptoms including skin rash, hair loss, tooth discoloration, halitosis reminiscent of garlic, gastrointestinal disturbances, numbness, paralysis, and hemiplegia [156,157].
Se deficiency, which affects about one billion people in the world, develops due to its insufficient consumption. This factor mainly depends on the geographical area and correlates with the low content of this microelement in the soil. The increased risk of mortality rates among patients with HF, development of type 2 diabetes mellitus, and heightened incidence of prostate cancer have been associated with Se deficiency [158,159,160]. This underscores the importance of maintaining Se intake within safe limits to avoid detrimental health consequences. Also, the diminished Se levels in the body are implicated in compromised fetal development, male infertility, and heightened susceptibility to asthma [130,161,162]. These effects stem from the attenuation of antioxidant defenses and the reduction in activity of glutathione peroxidase [126].
Furthermore, Se deficiency combined with coxsackievirus B3 infection leads to Keshan disease (KD), an endemic cardiomyopathy, reported in China [163]. KD was first identified in Keshan County in 1935, and due to its occurrence, the Chinese government introduced a Se supplementation program in the 1970s. Prior to the implementation of the program, adults residing in regions afflicted by KD demonstrated about 11 µg Se intakes a day [156]. It has been established that Se intakes of at least 20 mcg/day protect adults from KD [164]. The pathogenesis of KD is not yet fully understood, but the presence of infection, which leads to oxidative stress, is also an important risk factor for its development. This hypothesis is supported by the study by Pei et al., which demonstrated that lower activity of selenoproteins is a component of the disease in patients with KD [165]. It seems that the most crucial etiologic factors considered are deficiency of Se in diet and environment, above others considered, including toxins, viral infections, and lack of vitamins or minerals. In the study by Zhang et al., performed on animal models, it was observed that low Se levels affect PINK1/Parkin-mediated mitochondrial autophagy pathway, which exacerbate damage to the myocardium in KD [166]. Yet, the main evidence in Se involvement are benefits in patients receiving sodium selenite supplementation in endemic areas [138]. The clinical presentations observed in patients with KD includes congestive HF, cardiogenic shock, and arrhythmias [163]. In the imaging exam, dilatation of the heart cavities is observed, as well as numerous necrotic foci with various degrees of fibrosis [167].

Selenium Supplementation

Selenoproteins play a crucial role in preventing oxidative modification, thereby reducing inflammation and inhibiting platelet aggregation (Table 1). Due to these functions, it has been suggested that Se supplementation could reduce the risk of CVD and its risk of mortality. The meta-analysis of 16 prospective observational studies revealed a significant beneficial range of Se concentration between 55 and 145 μg/L, which was related to a lower risk of CVD development [168]. However, none of the nine randomized controlled trials indicated any overall effect of Se on CVD risk.
The necessity of adding Se to the antioxidant mix is also confirmed by another meta-analysis that analyzed 43 randomized controlled trials [101]. This study revealed a reduced risk for all-cause mortality when Se was part of the antioxidant mix (RR: 0.90; 95% CI: 0.82, 0.98; p = 0.02). Conversely, an antioxidant mix without Se showed indications of increased risk (RR: 1.09; 95% CI: 1.04, 1.13; p = 0.0002; I2 = 0%) for all-cause mortality. This research highlights the necessity of integrating Se into any antioxidant supplement mix to optimize its efficacy and mitigate potential CVD or all-cause mortality risk.
Another meta-analysis of 16 randomized controlled trials, involving 43,998 participants, studied the effect of Se supplementation on CHD [54]. The results of this study showed significant effects of Se supplementation on lowering serum CRP and increasing GSH-PX. However, there was no statistically significant association between Se supplementation and CHD mortality or improving their lipid profiles. Also, in another meta-analysis, to which patients were subjected to various micronutrient supplementation, Se had a neutral effect on CVD; its supplementation showed no therapeutic effects on lipid profile, blood pressure, myocardial infarction, stroke, or coronary heart disease [169].
Overall, the current clinical evidence does not support the routine use of Se supplements for preventing or treating heart disease, especially among patients who already maintain adequate serum Se levels. Further clinical trials are necessary to evaluate the contributions of dietary Se intake and supplementation in patients with CVD.

4. Conclusions

In conclusion, the current literature suggests that Se plays a crucial role in myocardial tissue homeostasis and in the progression of CVD. It has been shown that low levels of Se correlate with the risk of developing AF, both pre- and post-operatively. Attempts at supplementation have not yielded statistically significant results, both in reducing AF incidence and CHD mortality. However, these results demonstrated notable effects on GPx activity and serum CRP levels, offering potential benefits in managing CHD risk factors. Supplementation of this micronutrient appears promising in the prevention and treatment of HF. Despite the clear links that Se has to CVD development, further research is still required to assess the clinical power of these findings.

Author Contributions

Conceptualization, E.M., B.F. and J.R.; methodology, K.L., L.B., K.K., W.M., M.M. and A.W.; validation, E.M., B.F. and J.R.; formal analysis, K.L., L.B., K.K., W.M., M.M. and A.W.; investigation, K.L., L.B., K.K., W.M., M.M. and A.W.; resources, E.M., B.F. and J.R.; data curation, E.M.; writing—original draft preparation, K.L., L.B., K.K., W.M., M.M., and A.W.; writing—review and editing, E.M.; visualization, K.L., L.B., K.K., W.M., M.M., and A.W.; supervision, E.M., B.F., and J.R.; project administration, E.M.; funding acquisition, B.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data used in this article are sourced from materials mentioned in the References section.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kyu, H.H.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Disability-Adjusted Life-Years (DALYs) for 359 Diseases and Injuries and Healthy Life Expectancy (HALE) for 195 Countries and Territories, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1859–1922. [Google Scholar] [CrossRef] [PubMed]
  2. Roth, G.A.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Age-Sex-Specific Mortality for 282 Causes of Death in 195 Countries and Territories, 1980–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef] [PubMed]
  3. Amini, M.; Zayeri, F.; Salehi, M. Trend Analysis of Cardiovascular Disease Mortality, Incidence, and Mortality-to-Incidence Ratio: Results from Global Burden of Disease Study 2017. BMC Public Health 2021, 21, 401. [Google Scholar] [CrossRef] [PubMed]
  4. Senoner, T.; Dichtl, W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients 2019, 11, 2090. [Google Scholar] [CrossRef]
  5. Dubois-Deruy, E.; Peugnet, V.; Turkieh, A.; Pinet, F. Oxidative Stress in Cardiovascular Diseases. Antioxidants 2020, 9, 864. [Google Scholar] [CrossRef] [PubMed]
  6. Barchielli, G.; Capperucci, A.; Tanini, D. The Role of Selenium in Pathologies: An Updated Review. Antioxidants 2022, 11, 251. [Google Scholar] [CrossRef]
  7. Rayman, M.P. Selenium and Human Health. Lancet 2012, 379, 1256–1268. [Google Scholar] [CrossRef] [PubMed]
  8. Kuršvietienė, L.; Mongirdienė, A.; Bernatonienė, J.; Šulinskienė, J.; Stanevičienė, I. Selenium Anticancer Properties and Impact on Cellular Redox Status. Antioxidants 2020, 9, 80. [Google Scholar] [CrossRef] [PubMed]
  9. Konstantinidis, K.; Whelan, R.S.; Kitsis, R.N. Mechanisms of cell death in heart disease. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1552–1562. [Google Scholar] [CrossRef]
  10. Zhang, L.; Gao, Y.; Feng, H.; Zou, N.; Wang, K.; Sun, D. Effects of selenium deficiency and low protein intake on the apoptosis through a mitochondria-dependent pathway. J. Trace Elem. Med. Biol. 2019, 56, 21–30. [Google Scholar] [CrossRef]
  11. Zhang, C.; Deng, Y.; Lei, Y.; Zhao, J.; Wei, W.; Li, Y. Effects of selenium on myocardial apoptosis by modifying the activity of mitochondrial STAT3 and regulating potassium channel expression. Exp. Ther. Med. 2017, 14, 2201–2205. [Google Scholar] [CrossRef] [PubMed]
  12. Boengler, K.; Hilfiker-Kleiner, D.; Drexler, H.; Heusch, G.; Schulz, R. The myocardial JAK/STAT pathway: From protection to failure. Pharmacol. Ther. 2008, 120, 172–185. [Google Scholar] [CrossRef] [PubMed]
  13. Steinbrenner, H.; Bilgic, E.; Pinto, A.; Engels, M.; Wollschläger, L.; Döhrn, L.; Kellermann, K.; Boeken, U.; Akhyari, P.; Lichtenberg, A. Selenium Pretreatment for Mitigation of Ischemia/Reperfusion Injury in Cardiovascular Surgery: Influence on Acute Organ Damage and Inflammatory Response. Inflammation 2016, 39, 1363–1376. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, F.; Li, X.; Wei, Y. Selenium and Selenoproteins in Health. Biomolecules 2023, 13, 799. [Google Scholar] [CrossRef] [PubMed]
  15. Kryukov, G.V.; Castellano, S.; Novoselov, S.V.; Lobanov, A.V.; Zehtab, O.; Guigó, R.; Gladyshev, V.N. Characterization of Mammalian Selenoproteomes. Science 2003, 300, 1439–1443. [Google Scholar] [CrossRef] [PubMed]
  16. Peeler, J.C.; Weerapana, E. Chemical Biology Approaches to Interrogate the Selenoproteome. Acc. Chem. Res. 2019, 52, 2832–2840. [Google Scholar] [CrossRef] [PubMed]
  17. Benstoem, C.; Goetzenich, A.; Kraemer, S.; Borosch, S.; Manzanares, W.; Hardy, G.; Stoppe, C. Selenium and Its Supplementation in Cardiovascular Disease—What Do We Know? Nutrients 2015, 7, 3094–3118. [Google Scholar] [CrossRef] [PubMed]
  18. Ren, B.; Liu, M.; Ni, J.; Tian, J. Role of Selenoprotein F in Protein Folding and Secretion: Potential Involvement in Human Disease. Nutrients 2018, 10, 1619. [Google Scholar] [CrossRef] [PubMed]
  19. Cox, A.G.; Tsomides, A.; Kim, A.J.; Saunders, D.; Hwang, K.L.; Evason, K.J.; Heidel, J.; Brown, K.K.; Yuan, M.; Lien, E.C.; et al. Selenoprotein H Is an Essential Regulator of Redox Homeostasis That Cooperates with P53 in Development and Tumorigenesis. Proc. Natl. Acad. Sci. USA 2016, 113, E5562–E5571. [Google Scholar] [CrossRef]
  20. Wu, R.T.Y.; Cao, L.; Chen, B.P.C.; Cheng, W.-H. Selenoprotein H Suppresses Cellular Senescence through Genome Maintenance and Redox Regulation. J. Biol. Chem. 2014, 289, 34378–34388. [Google Scholar] [CrossRef]
  21. Horibata, Y.; Elpeleg, O.; Eran, A.; Hirabayashi, Y.; Savitzki, D.; Tal, G.; Mandel, H.; Sugimoto, H. EPT1 (Selenoprotein I) Is Critical for the Neural Development and Maintenance of Plasmalogen in Humans. J. Lipid Res. 2018, 59, 1015–1026. [Google Scholar] [CrossRef] [PubMed]
  22. Ma, C.; Martinez-Rodriguez, V.; Hoffmann, P.R. Roles for Selenoprotein I and Ethanolamine Phospholipid Synthesis in T Cell Activation. Int. J. Mol. Sci. 2021, 22, 11174. [Google Scholar] [CrossRef] [PubMed]
  23. Fredericks, G.J.; Hoffmann, P.R. Selenoprotein K and Protein Palmitoylation. Antioxid. Redox Signal. 2015, 23, 854–862. [Google Scholar] [CrossRef] [PubMed]
  24. Lu, C.; Qiu, F.; Zhou, H.; Peng, Y.; Hao, W.; Xu, J.; Yuan, J.; Wang, S.; Qiang, B.; Xu, C.; et al. Identification and Characterization of Selenoprotein K: An Antioxidant in Cardiomyocytes. FEBS Lett. 2006, 580, 5189–5197. [Google Scholar] [CrossRef] [PubMed]
  25. Jia, S.-Z.; Xu, X.-W.; Zhang, Z.-H.; Chen, C.; Chen, Y.-B.; Huang, S.-L.; Liu, Q.; Hoffmann, P.R.; Song, G.-L. Selenoprotein K Deficiency-Induced Apoptosis: A Role for Calpain and the ERS Pathway. Redox Biol. 2021, 47, 102154. [Google Scholar] [CrossRef] [PubMed]
  26. Reeves, M.A.; Bellinger, F.P.; Berry, M.J. The Neuroprotective Functions of Selenoprotein M and Its Role in Cytosolic Calcium Regulation. Antioxid. Redox Signal. 2010, 12, 809–818. [Google Scholar] [CrossRef]
  27. Nunes, L.G.A.; Cain, A.; Comyns, C.; Hoffmann, P.R.; Krahn, N. Deciphering the Role of Selenoprotein M. Antioxidants 2023, 12, 1906. [Google Scholar] [CrossRef] [PubMed]
  28. Castets, P.; Bertrand, A.T.; Beuvin, M.; Ferry, A.; Le Grand, F.; Castets, M.; Chazot, G.; Rederstorff, M.; Krol, A.; Lescure, A.; et al. Satellite Cell Loss and Impaired Muscle Regeneration in Selenoprotein N Deficiency. Hum. Mol. Genet. 2011, 20, 694–704. [Google Scholar] [CrossRef] [PubMed]
  29. Zito, E.; Ferreiro, A. Calcium and Redox Liaison: A Key Role of Selenoprotein N in Skeletal Muscle. Cells 2021, 10, 1116. [Google Scholar] [CrossRef]
  30. Sreelatha, A.; Yee, S.S.; Lopez, V.A.; Park, B.C.; Kinch, L.N.; Pilch, S.; Servage, K.A.; Zhang, J.; Jiou, J.; Karasiewicz-Urbańska, M.; et al. Protein AMPylation by an Evolutionarily Conserved Pseudokinase. Cell 2018, 175, 809–821.e19. [Google Scholar] [CrossRef]
  31. Schomburg, L.; Orho-Melander, M.; Struck, J.; Bergmann, A.; Melander, O. Selenoprotein-P Deficiency Predicts Cardiovascular Disease and Death. Nutrients 2019, 11, 1852. [Google Scholar] [CrossRef]
  32. Schweizer, U.; Wirth, E.K.; Klopstock, T.; Hölter, S.M.; Becker, L.; Moskovitz, J.; Grune, T.; Fuchs, H.; Gailus-Durner, V.; Hrabe De Angelis, M.; et al. Seizures, Ataxia and Parvalbumin-Expressing Interneurons Respond to Selenium Supply in Selenop-Deficient Mice. Redox Biol. 2022, 57, 102490. [Google Scholar] [CrossRef]
  33. Ekoue, D.N.; Ansong, E.; Liu, L.; Macias, V.; Deaton, R.; Lacher, C.; Picklo, M.; Nonn, L.; Gann, P.H.; Kajdacsy-Balla, A.; et al. Correlations of SELENOF and SELENOP Genotypes with Serum Selenium Levels and Prostate Cancer. Prostate 2018, 78, 279–288. [Google Scholar] [CrossRef] [PubMed]
  34. Jia, Y.; Zhou, J.; Liu, H.; Huang, K. Effect of Methionine Sulfoxide Reductase B1 (SelR) Gene Silencing on Peroxynitrite-Induced F-Actin Disruption in Human Lens Epithelial Cells. Biochem. Biophys. Res. Commun. 2014, 443, 876–881. [Google Scholar] [CrossRef] [PubMed]
  35. Santos, L.R.; Durães, C.; Ziros, P.G.; Pestana, A.; Esteves, C.; Neves, C.; Carvalho, D.; Bongiovanni, M.; Renaud, C.O.; Chartoumpekis, D.V.; et al. Interaction of Genetic Variations in NFE2L2 and SELENOS Modulates the Risk of Hashimoto’s Thyroiditis. Thyroid 2019, 29, 1302–1315. [Google Scholar] [CrossRef] [PubMed]
  36. Strauss, E.; Tomczak, J.; Staniszewski, R.; Oszkinis, G. Associations and Interactions between Variants in Selenoprotein Genes, Selenoprotein Levels and the Development of Abdominal Aortic Aneurysm, Peripheral Arterial Disease, and Heart Failure. PLoS ONE 2018, 13, e0203350. [Google Scholar] [CrossRef] [PubMed]
  37. Anouar, Y.; Lihrmann, I.; Falluel-Morel, A.; Boukhzar, L. Selenoprotein T Is a Key Player in ER Proteostasis, Endocrine Homeostasis and Neuroprotection. Free Radic. Biol. Med. 2018, 127, 145–152. [Google Scholar] [CrossRef]
  38. Rocca, C.; Boukhzar, L.; Granieri, M.C.; Alsharif, I.; Mazza, R.; Lefranc, B.; Tota, B.; Leprince, J.; Cerra, M.C.; Anouar, Y.; et al. A Selenoprotein T-derived Peptide Protects the Heart against Ischaemia/Reperfusion Injury through Inhibition of Apoptosis and Oxidative Stress. Acta Physiol. 2018, 223, e13067. [Google Scholar] [CrossRef] [PubMed]
  39. Dogaru, C.B.; Duță, C.; Muscurel, C.; Stoian, I. “Alphabet” Selenoproteins: Implications in Pathology. Int. J. Mol. Sci. 2023, 24, 15344. [Google Scholar] [CrossRef]
  40. Kim, H.; Lee, K.; Kim, J.M.; Kim, M.Y.; Kim, J.-R.; Lee, H.-W.; Chung, Y.W.; Shin, H.-I.; Kim, T.; Park, E.-S.; et al. Selenoprotein W Ensures Physiological Bone Remodeling by Preventing Hyperactivity of Osteoclasts. Nat. Commun. 2021, 12, 2258. [Google Scholar] [CrossRef]
  41. Liao, C.; Hardison, R.C.; Kennett, M.J.; Carlson, B.A.; Paulson, R.F.; Prabhu, K.S. Selenoproteins Regulate Stress Erythroid Progenitors and Spleen Microenvironment during Stress Erythropoiesis. Blood 2018, 131, 2568–2580. [Google Scholar] [CrossRef] [PubMed]
  42. Qi, Z.; Duan, A.; Ng, K. Selenoproteins in Health. Molecules 2023, 29, 136. [Google Scholar] [CrossRef] [PubMed]
  43. Metere, A.; Frezzotti, F.; Graves, C.E.; Vergine, M.; De Luca, A.; Pietraforte, D.; Giacomelli, L. A possible role for selenoprotein glutathione peroxidase (GPx1) and thioredoxin reductases (TrxR1) in thyroid cancer: Our experience in thyroid surgery. Cancer Cell Int. 2018, 18, 7. [Google Scholar] [CrossRef] [PubMed]
  44. Arnér, E.S.J. Targeting the selenoprotein thioredoxin reductase 1 for anticancer therapy. Adv. Cancer Res. 2017, 136, 139–151. [Google Scholar] [CrossRef] [PubMed]
  45. Jiang, N.; Liu, J.; Guan, C.; Ma, C.; An, J.; Tang, X. Thioredoxin-Interacting Protein: A New Therapeutic Target in Bone Metabolism Disorders? Front. Immunol. 2022, 13, 955128. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, S.; Kim, S.M.; Lee, R.T. Thioredoxin and Thioredoxin Target Proteins: From Molecular Mechanisms to Functional Significance. Antioxid. Redox Signal. 2013, 18, 1165–1207. [Google Scholar] [CrossRef] [PubMed]
  47. Maeda, T.; Van Hooser, J.P.; Driessen, C.A.G.G.; Filipek, S.; Janssen, J.J.M.; Palczewski, K. Evaluation of the Role of the Retinal G Protein-coupled Receptor (RGR) in the Vertebrate Retina in Vivo. J. Neurochem. 2003, 85, 944–956. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, P.; Hao, W.; Rife, L.; Wang, X.P.; Shen, D.; Chen, J.; Ogden, T.; Van Boemel, G.B.; Wu, L.; Yang, M.; et al. A Photic Visual Cycle of Rhodopsin Regeneration Is Dependent on Rgr. Nat. Genet. 2001, 28, 256–260. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, J.-X.; Wang, Z.-M.; Zhang, J.-J.; Zhu, L.-L.; Gao, X.-F.; Chen, S.-L. Association of Glutathione Peroxidase-1 (GPx-1) Rs1050450 Pro198Leu and Pro197Leu Polymorphisms with Cardiovascular Risk: A Meta-Analysis of Observational Studies. J. Geriatr. Cardiol. 2014, 11, 141–150. [Google Scholar] [CrossRef]
  50. Da Rocha, T.J.; Silva Alves, M.; Guisso, C.C.; De Andrade, F.M.; Camozzato, A.; De Oliveira, A.A.; Fiegenbaum, M. Association of GPX1 and GPX4 Polymorphisms with Episodic Memory and Alzheimer’s Disease. Neurosci. Lett. 2018, 666, 32–37. [Google Scholar] [CrossRef]
  51. Liu, D.; Liu, L.; Hu, Z.; Song, Z.; Wang, Y.; Chen, Z. Evaluation of the Oxidative Stress–Related Genes ALOX5, ALOX5AP, GPX1, GPX3 and MPO for Contribution to the Risk of Type 2 Diabetes Mellitus in the Han Chinese Population. Diabetes Vasc. Dis. Res. 2018, 15, 336–339. [Google Scholar] [CrossRef] [PubMed]
  52. Hu, Y.J.; Diamond, A.M. Role of Glutathione Peroxidase 1 in Breast Cancer: Loss of Heterozygosity and Allelic Differences in the Response to Selenium. Cancer Res. 2003, 63, 3347–3351. [Google Scholar] [PubMed]
  53. Handy, D.E.; Loscalzo, J. The Role of Glutathione Peroxidase-1 in Health and Disease. Free Radic. Biol. Med. 2022, 188, 146–161. [Google Scholar] [CrossRef] [PubMed]
  54. Esworthy, R.S.; Doroshow, J.H.; Chu, F.-F. The Beginning of GPX2 and 30 Years Later. Free Radic. Biol. Med. 2022, 188, 419–433. [Google Scholar] [CrossRef] [PubMed]
  55. Kipp, A.P.; Müller, M.F.; Göken, E.M.; Deubel, S.; Brigelius-Flohé, R. The Selenoproteins GPx2, TrxR2 and TrxR3 Are Regulated by WntSignalling in the Intestinal Epithelium. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2012, 1820, 1588–1596. [Google Scholar] [CrossRef] [PubMed]
  56. Chang, C.; Worley, B.L.; Phaëton, R.; Hempel, N. Extracellular Glutathione Peroxidase GPx3 and Its Role in Cancer. Cancers 2020, 12, 2197. [Google Scholar] [CrossRef] [PubMed]
  57. Cardoso, B.R.; Hare, D.J.; Bush, A.I.; Roberts, B.R. Glutathione Peroxidase 4: A New Player in Neurodegeneration? Mol. Psychiatry 2017, 22, 328–335. [Google Scholar] [CrossRef] [PubMed]
  58. Maiorino, M.; Bosello, V.; Ursini, F.; Foresta, C.; Garolla, A.; Scapin, M.; Sztajer, H.; Flohé, L. Genetic Variations of Gpx-4 and Male Infertility in Humans1. Biol. Reprod. 2003, 68, 1134–1141. [Google Scholar] [CrossRef] [PubMed]
  59. Li, P.; Jiang, M.; Li, K.; Li, H.; Zhou, Y.; Xiao, X.; Xu, Y.; Krishfield, S.; Lipsky, P.E.; Tsokos, G.C.; et al. Glutathione Peroxidase 4–Regulated Neutrophil Ferroptosis Induces Systemic Autoimmunity. Nat. Immunol. 2021, 22, 1107–1117. [Google Scholar] [CrossRef]
  60. Pei, J.; Pan, X.; Wei, G.; Hua, Y. Research Progress of Glutathione Peroxidase Family (GPX) in Redoxidation. Front. Pharmacol. 2023, 14, 1147414. [Google Scholar] [CrossRef]
  61. Kieliszek, M.; Bano, I. Selenium as an Important Factor in Various Disease States—A Review. EXCLI J. 2022, 21, 948–966. [Google Scholar] [CrossRef] [PubMed]
  62. Forouzanfar, M.H.; Liu, P.; Roth, G.A.; Ng, M.; Biryukov, S.; Marczak, L.; Alexander, L.; Estep, K.; Hassen Abate, K.; Akinyemiju, T.F.; et al. Global Burden of Hypertension and Systolic Blood Pressure of at Least 110 to 115 Mm Hg, 1990–2015. JAMA 2017, 317, 165. [Google Scholar] [CrossRef] [PubMed]
  63. Whelton, P.K.; Carey, R.M.; Mancia, G.; Kreutz, R.; Bundy, J.D.; Williams, B. Harmonization of the American College of Cardiology/American Heart Association and European Society of Cardiology/European Society of Hypertension Blood Pressure/Hypertension Guidelines: Comparisons, Reflections, and Recommendations. Circulation 2022, 146, 868–877. [Google Scholar] [CrossRef] [PubMed]
  64. Kario, K.; Harada, N.; Okura, A. Digital Therapeutics in Hypertension: Evidence and Perspectives. Hypertension 2022, 79, 2148–2158. [Google Scholar] [CrossRef] [PubMed]
  65. Hariharan, S.; Dharmaraj, S. Selenium and Selenoproteins: It’s Role in Regulation of Inflammation. Inflammopharmacology 2020, 28, 667–695. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, Z.; Zhao, L.; Zhou, X.; Meng, X.; Zhou, X. Role of Inflammation, Immunity, and Oxidative Stress in Hypertension: New Insights and Potential Therapeutic Targets. Front. Immunol. 2023, 13, 1098725. [Google Scholar] [CrossRef] [PubMed]
  67. Agita, A.; Alsagaff, M.T. Inflammation, Immunity, and Hypertension. Acta Med. Indones. 2017, 49, 158–165. [Google Scholar] [PubMed]
  68. Jayedi, A.; Rahimi, K.; Bautista, L.E.; Nazarzadeh, M.; Zargar, M.S.; Shab-Bidar, S. Inflammation Markers and Risk of Developing Hypertension: A Meta-Analysis of Cohort Studies. Heart 2019, 105, 686–692. [Google Scholar] [CrossRef]
  69. Gholizadeh, M.; Khalili, A.; Roodi, P.B.; Saeedy, S.A.G.; Najafi, S.; Keshavarz Mohammadian, M.; Djafarian, K. Selenium Supplementation Decreases CRP and IL-6 and Increases TNF-Alpha: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Trace Elem. Med. Biol. 2023, 79, 127199. [Google Scholar] [CrossRef]
  70. Shen, Y.; Dong, Z.; Fan, F.; Li, K.; Zhu, S.; Dai, R.; Huang, J.; Xie, N.; He, L.; Gong, Z.; et al. Targeting Cytokine-like Protein FAM3D Lowers Blood Pressure in Hypertension. Cell Rep. Med. 2023, 4, 101072. [Google Scholar] [CrossRef]
  71. Mazloomi, S.; Khodadadi, I.; Alimohammadi, S.; Shafiee, G. Correlation of Thioredoxin Reductase (TrxR) and Nitric Oxide Synthase (NOS) Activities with Serum Trace Elements in Preeclampsia. Clin. Exp. Hypertens. 2021, 43, 120–124. [Google Scholar] [CrossRef] [PubMed]
  72. Isselbacher, E.M.; Preventza, O.; Hamilton Black, J.; Augoustides, J.G.; Beck, A.W.; Bolen, M.A.; Braverman, A.C.; Bray, B.E.; Brown-Zimmerman, M.M.; Chen, E.P.; et al. 2022 ACC/AHA Guideline for the Diagnosis and Management of Aortic Disease: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. Circulation 2022, 146, 334–482. [Google Scholar] [CrossRef] [PubMed]
  73. Park, S.; Lakatta, E.G. Role of Inflammation in the Pathogenesis of Arterial Stiffness. Yonsei Med. J. 2012, 53, 258. [Google Scholar] [CrossRef] [PubMed]
  74. Bastola, M.M.; Locatis, C.; Maisiak, R.; Fontelo, P. Selenium, Copper, Zinc and Hypertension: An Analysis of the National Health and Nutrition Examination Survey (2011–2016). BMC Cardiovasc. Disord. 2020, 20, 45. [Google Scholar] [CrossRef] [PubMed]
  75. Mark, S.D.; Wang, W.; Fraumeni, J.F.; Li, J.Y.; Taylor, P.R.; Wang, G.Q.; Dawsey, S.M.; Li, B.; Blot, W.J. Do Nutritional Supplements Lower the Risk of Stroke or Hypertension? Epidemiology 1998, 9, 9–15. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, Y.; Yu, Z. Association between Dietary Selenium Intake and the Prevalence of Hypertension: Results from the National Health and Nutrition Examination Survey 2003–2018. Front. Immunol. 2024, 15, 1338745. [Google Scholar] [CrossRef] [PubMed]
  77. Lei, L.; Zhang, F.; Huang, J.; Yang, X.; Zhou, X.; Yan, H.; Chen, C.; Zheng, S.; Si, L.; Jose, P.A.; et al. Selenium Deficiency Causes Hypertension by Increasing Renal AT1 Receptor Expression via GPx1/H2O2/NF-κB Pathway. Free Radic. Biol. Med. 2023, 200, 59–72. [Google Scholar] [CrossRef] [PubMed]
  78. Tan, Q.; Huang, Y.; Liu, X.; Liu, L.; Lo, K.; Chen, J.; Feng, Y. A U-Shaped Relationship Between Selenium Concentrations and All-Cause or Cardiovascular Mortality in Patients with Hypertension. Front. Cardiovasc. Med. 2021, 8, 671618. [Google Scholar] [CrossRef]
  79. Torkzaban, A.; Naeini, A.A.; Hassanzadeh, A.; Namdari, M. The Relationship between Serum Vitamin C and Uric Acid Levels, Antioxidant Status and Coronary Artery Disease: A Case-Control Study. Clin. Nutr. Res. 2020, 9, 307. [Google Scholar] [CrossRef]
  80. Huang, H.; Wang, Y.; Wang, X.; Lei, Y. Association of CYP4F2 and CTRP9 Polymorphisms and Serum Selenium Levels with Coronary Artery Disease. Medicine 2020, 99, e20494. [Google Scholar] [CrossRef]
  81. Frączek-Jucha, M.; Szlósarczyk, B.; Kabat, M.; Czubek, U.; Nessler, J.; Gackowski, A. Low Triiodothyronine Syndrome and Serum Selenium Status in the Course of Acute Myocardial Infarction. Pol. Merkur. Lek. 2019, 47, 45–51. [Google Scholar]
  82. Hu, L.; Shi, Q.; Shi, M.; Liu, R.; Wang, C. Diagnostic Value of PCT and CRP for Detecting Serious Bacterial Infections in Patients with Fever of Unknown Origin: A Systematic Review and Meta-Analysis. Appl. Immunohistochem. Mol. Morphol. 2017, 25, e61–e69. [Google Scholar] [CrossRef]
  83. Kok, F.J.; Hofman, A.; Witteman, J.C.; de Bruijn, A.M.; Kruyssen, D.H.; de Bruin, M.; Valkenburg, H.A. Decreased Selenium Levels in Acute Myocardial Infarction. JAMA 1989, 261, 1161–1164. [Google Scholar] [CrossRef]
  84. Kutil, B.; Ostadal, P.; Vejvoda, J.; Kukacka, J.; Cepova, J.; Alan, D.; Krüger, A.; Vondrakova, D. Alterations in Serum Selenium Levels and Their Relation to Troponin I in Acute Myocardial Infarction. Mol. Cell Biochem. 2010, 345, 23–27. [Google Scholar] [CrossRef]
  85. Venardos, K.; Harrison, G.; Headrick, J.; Perkins, A. Selenium Supplementation and Ischemia–Reperfusion Injury in Rats. Redox Rep. 2004, 9, 317–320. [Google Scholar] [CrossRef]
  86. Zachara, B.A.; Ukleja-Adamowicz, M.; Nartowicz, E.; Łecka, J. Increased Plasma Glutathione Peroxidase Activity in Patients with Acute Myocardial Infarction. Med. Sci. Monit. 2001, 7, 415–420. [Google Scholar]
  87. Flores-Mateo, G.; Navas-Acien, A.; Pastor-Barriuso, R.; Guallar, E. Selenium and Coronary Heart Disease: A Meta-Analysis. Am. J. Clin. Nutr. 2006, 84, 762–773. [Google Scholar] [CrossRef]
  88. Kuria, A.; Tian, H.; Li, M.; Wang, Y.; Aaseth, J.O.; Zang, J.; Cao, Y. Selenium Status in the Body and Cardiovascular Disease: A Systematic Review and Meta-Analysis. Crit. Rev. Food Sci. Nutr. 2021, 61, 3616–3625. [Google Scholar] [CrossRef]
  89. Yuan, Y.; Xiao, Y.; Feng, W.; Liu, Y.; Yu, Y.; Zhou, L.; Qiu, G.; Wang, H.; Liu, B.; Liu, K.; et al. Plasma Metal Concentrations and Incident Coronary Heart Disease in Chinese Adults: The Dongfeng-Tongji Cohort. Environ. Health Perspect. 2017, 125, 107007. [Google Scholar] [CrossRef]
  90. Xie, B.; Wang, J.; Zhang, J.; Chen, M. Dietary and Serum Selenium in Coronary Heart Disease and All-Cause Mortality: An International Perspective. Asia Pac. J. Clin. Nutr. 2020, 29, 827–838. [Google Scholar] [CrossRef] [PubMed]
  91. Rath, A.A.; Lam, H.S.; Schooling, C.M. Effects of Selenium on Coronary Artery Disease, Type 2 Diabetes and Their Risk Factors: A Mendelian Randomization Study. Eur. J. Clin. Nutr. 2021, 75, 1668–1678. [Google Scholar] [CrossRef] [PubMed]
  92. Xiang, S.; Dai, Z.; Man, C.; Fan, Y. Circulating Selenium and Cardiovascular or All-Cause Mortality in the General Population: A Meta-Analysis. Biol. Trace Elem. Res. 2020, 195, 55–62. [Google Scholar] [CrossRef] [PubMed]
  93. Bleys, J.; Navas-Acien, A.; Guallar, E. Serum Selenium Levels and All-Cause, Cancer, and Cardiovascular Mortality among US Adults. Arch. Intern. Med. 2008, 168, 404–410. [Google Scholar] [CrossRef] [PubMed]
  94. Fairweather-Tait, S.J.; Bao, Y.; Broadley, M.R.; Collings, R.; Ford, D.; Hesketh, J.E.; Hurst, R. Selenium in Human Health and Disease. Antioxid. Redox Signal. 2011, 14, 1337–1383. [Google Scholar] [CrossRef] [PubMed]
  95. Xie, Y.-H.; Chen, Y.-X.; Fang, J.-Y. Comprehensive Review of Targeted Therapy for Colorectal Cancer. Signal Transduct. Target Ther. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed]
  96. Ju, W.; Li, X.; Li, Z.; Wu, G.R.; Fu, X.F.; Yang, X.M.; Zhang, X.Q.; Gao, X.B. The Effect of Selenium Supplementation on Coronary Heart Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Trace Elem. Med. Biol. 2017, 44, 8–16. [Google Scholar] [CrossRef] [PubMed]
  97. Schnabel, R.; Lubos, E.; Messow, C.M.; Sinning, C.R.; Zeller, T.; Wild, P.S.; Peetz, D.; Handy, D.E.; Munzel, T.; Loscalzo, J.; et al. Selenium Supplementation Improves Antioxidant Capacity in Vitro and in Vivo in Patients with Coronary Artery Disease. Am. Heart J. 2008, 156, 1201.e1–1201.e11. [Google Scholar] [CrossRef] [PubMed]
  98. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the Diagnosis and Treatment of Acute and Chronic Heart Failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  99. Mirdamadi, A.; Rafiei, R.; Kahazaipour, G.; Fouladi, L. Selenium Level in Patients with Heart Failure versus Normal Individuals. Int. J. Prev. Med. 2019, 10, 210. [Google Scholar] [CrossRef]
  100. Bomer, N.; Grote Beverborg, N.; Hoes, M.F.; Streng, K.W.; Vermeer, M.; Dokter, M.M.; IJmker, J.; Anker, S.D.; Cleland, J.G.F.; Hillege, H.L.; et al. Selenium and Outcome in Heart Failure. Eur. J. Heart Fail. 2020, 22, 1415–1423. [Google Scholar] [CrossRef]
  101. Jenkins, D.J.; Kitts, D.; Giovannucci, E.L.; Sahye-Pudaruth, S.; Paquette, M.; Blanco Mejia, S.; Patel, D.; Kavanagh, M.; Tsirakis, T.; Kendall, C.W.; et al. Selenium, Antioxidants, Cardiovascular Disease, and All-Cause Mortality: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am. J. Clin. Nutr. 2020, 112, 1642–1652. [Google Scholar] [CrossRef]
  102. Staerk, L.; Sherer, J.A.; Ko, D.; Benjamin, E.J.; Helm, R.H. Atrial Fibrillation: Epidemiology, Pathophysiology, and Clinical Outcomes. Circ. Res. 2017, 120, 1501–1517. [Google Scholar] [CrossRef]
  103. Sagris, M.; Vardas, E.P.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Tousoulis, D. Atrial Fibrillation: Pathogenesis, Predisposing Factors, and Genetics. Int. J. Mol. Sci. 2021, 23, 6. [Google Scholar] [CrossRef]
  104. Sovari, A.A.; Dudley, S.C. Reactive Oxygen Species-Targeted Therapeutic Interventions for Atrial Fibrillation. Front. Physiol. 2012, 3, 311. [Google Scholar] [CrossRef]
  105. Brouwers, F.P.; De Boer, R.A.; Van Der Harst, P.; Voors, A.A.; Gansevoort, R.T.; Bakker, S.J.; Hillege, H.L.; Van Veldhuisen, D.J.; Van Gilst, W.H. Incidence and Epidemiology of New Onset Heart Failure with Preserved vs. Reduced Ejection Fraction in a Community-Based Cohort: 11-Year Follow-up of PREVEND. Eur. Heart J. 2013, 34, 1424–1431. [Google Scholar] [CrossRef]
  106. Al-Mubarak, A.A.; Grote Beverborg, N.; Zwartkruis, V.; Van Deutekom, C.; De Borst, M.H.; Gansevoort, R.T.; Bakker, S.J.L.; Touw, D.J.; De Boer, R.A.; Van Der Meer, P.; et al. Micronutrient Deficiencies and New-Onset Atrial Fibrillation in a Community-Based Cohort: Data from PREVEND. Clin. Res. Cardiol. 2023. [Google Scholar] [CrossRef]
  107. Ardahanli, I.; Ozkan, H.I. Comparison of Serum Selenium Levels Between Patients with Newly Diagnosed Atrial Fibrillation and Normal Controls. Biol. Trace Elem. Res. 2022, 200, 3925–3931. [Google Scholar] [CrossRef]
  108. Bazargani, Z.; Shojaeifard, M.B.; Valizadeh, G.A. Comparison of Trace Elements Levels in Atrial Fibrillation Patients and Healthy Group. J. Pharm. Res. Int. 2020, 32, 113–119. [Google Scholar] [CrossRef]
  109. Negreva, M.; Georgiev, S.; Vitlianova, K.; Georgieva, R. Selenium Status in Paroxysmal Atrial Fibrillation. Int. J. Med. Pharm. 2015, 3, 17–26. [Google Scholar] [CrossRef]
  110. Samman Tahhan, A.; Sandesara, P.B.; Hayek, S.S.; Alkhoder, A.; Chivukula, K.; Hammadah, M.; Mohamed-Kelli, H.; O’Neal, W.T.; Topel, M.; Ghasemzadeh, N.; et al. Association between Oxidative Stress and Atrial Fibrillation. Heart Rhythm 2017, 14, 1849–1855. [Google Scholar] [CrossRef] [PubMed]
  111. Mirhoseini, M.F.; Hamblin, S.E.; Moore, W.P.; Pouliot, J.; Jenkins, J.M.; Wang, W.; Chandrasekhar, R.; Collier, B.R.; Patel, M.B. Antioxidant Supplementation and Atrial Arrhythmias in Critically Ill Trauma Patients. J. Surg. Res. 2018, 222, 10–16. [Google Scholar] [CrossRef]
  112. Szyller, J.; Jagielski, D.; Bil-Lula, I. Antioxidants in Arrhythmia Treatment—Still a Controversy? A Review of Selected Clinical and Laboratory Research. Antioxidants 2022, 11, 1109. [Google Scholar] [CrossRef]
  113. Peretto, G.; Durante, A.; Limite, L.R.; Cianflone, D. Postoperative Arrhythmias after Cardiac Surgery: Incidence, Risk Factors, and Therapeutic Management. Cardiol. Res. Pract. 2014, 2014, 1–15. [Google Scholar] [CrossRef]
  114. McDonald, C.; Fraser, J.; Shekar, K.; Clarke, A.; Coombes, J.; Barnett, A.; Pearse, B.; Fung, L. Low Preoperative Selenium Is Associated with Post-Operative Atrial Fibrillation in Patients Having Intermediate-Risk Coronary Artery Surgery. Eur. J. Clin. Nutr. 2016, 70, 1138–1143. [Google Scholar] [CrossRef]
  115. Kyaruzi, M.; Iyigün, T.; Diker, V.O.; Kurt, B.O.; Kahraman, Z.; Onan, B. Trace Element Status and Postoperative Morbidity After On-Pump Coronary Artery Bypass Surgery. Biol. Trace Elem. Res. 2023, 201, 2711–2720. [Google Scholar] [CrossRef]
  116. Koszta, G.; Kacska, Z.; Szatmári, K.; Szerafin, T.; Fülesdi, B. Lower Whole Blood Selenium Level Is Associated with Higher Operative Risk and Mortality Following Cardiac Surgery. J. Anesth. 2012, 26, 812–821. [Google Scholar] [CrossRef]
  117. Stoppe, C.; Schälte, G.; Rossaint, R.; Coburn, M.; Graf, B.; Spillner, J.; Marx, G.; Rex, S. The Intraoperative Decrease of Selenium Is Associated with the Postoperative Development of Multiorgan Dysfunction in Cardiac Surgical Patients*. Crit. Care Med. 2011, 39, 1879–1885. [Google Scholar] [CrossRef]
  118. Zhou, J.-Y.; Hou, H.-T.; Wang, S.-F.; Yang, Q.; He, G.-W. Plasma Concentrations of Trace Elements Selenium and Cobalt During and After Coronary Artery Bypass Grafting Surgery. Acta Med. Okayama 2022, 76, 33–39. [Google Scholar] [CrossRef]
  119. Eslami, G.; Salehi, R.; Samaee, H.; Habibi, V.; Shokrzadeh, M.; Moradimajd, P. Relationship Between Selenium Trace and Patient Outcome After Open-Heart Surgery. Anesth. Pain Med. 2020, 10, e105895. [Google Scholar] [CrossRef]
  120. Stevanovic, A.; Coburn, M.; Menon, A.; Rossaint, R.; Heyland, D.; Schälte, G.; Werker, T.; Wonisch, W.; Kiehntopf, M.; Goetzenich, A.; et al. The Importance of Intraoperative Selenium Blood Levels on Organ Dysfunction in Patients Undergoing Off-Pump Cardiac Surgery: A Randomised Controlled Trial. PLoS ONE 2014, 9, e104222. [Google Scholar] [CrossRef]
  121. Rehan, S.T.; Hussain, H.U.; Imran, L.; Eqbal, F.; Asghar, M.S. Perioperative Selenium Administration in Cardiac Surgery Patients, a Way out to Reduce Post Surgical Adversities? A Meta Analysis. Front. Cardiovasc. Med. 2023, 10, 1235247. [Google Scholar] [CrossRef]
  122. Stoppe, C.; McDonald, B.; Meybohm, P.; Christopher, K.B.; Fremes, S.; Whitlock, R.; Mohammadi, S.; Kalavrouziotis, D.; Elke, G.; Rossaint, R.; et al. Effect of High-Dose Selenium on Postoperative Organ Dysfunction and Mortality in Cardiac Surgery Patients: The SUSTAIN CSX Randomized Clinical Trial. JAMA Surg. 2023, 158, 235. [Google Scholar] [CrossRef]
  123. Schmidt, T.; Pargger, H.; Seeberger, E.; Eckhart, F.; Von Felten, S.; Haberthür, C. Effect of High-Dose Sodium Selenite in Cardiac Surgery Patients: A Randomized Controlled Bi-Center Trial. Clin. Nutr. 2018, 37, 1172–1180. [Google Scholar] [CrossRef]
  124. Kamali, A.; Amirani, E.; Asemi, Z. Effects of Selenium Supplementation on Metabolic Status in Patients Undergoing for Coronary Artery Bypass Grafting (CABG) Surgery: A Randomized, Double-Blind, Placebo-Controlled Trial. Biol. Trace Elem. Res. 2019, 191, 331–337. [Google Scholar] [CrossRef]
  125. Ibrahim, S.A.Z.; Kerkadi, A.; Agouni, A. Selenium and Health: An Update on the Situation in the Middle East and North Africa. Nutrients 2019, 11, 1457. [Google Scholar] [CrossRef]
  126. Genchi, G.; Lauria, G.; Catalano, A.; Sinicropi, M.S.; Carocci, A. Biological Activity of Selenium and Its Impact on Human Health. Int. J. Mol. Sci. 2023, 24, 2633. [Google Scholar] [CrossRef]
  127. EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA); Turck, D.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.; Kearney, J.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Scientific Opinion on the Safety of Selenite Triglycerides as a Source of Selenium Added for Nutritional Purposes to Food Supplements. EFSA J. 2020, 18, e06134. [Google Scholar] [CrossRef]
  128. Kieliszek, M.; Błażejak, S. Selenium: Significance, and Outlook for Supplementation. Nutrition 2013, 29, 713–718. [Google Scholar] [CrossRef]
  129. Galan-Chilet, I.; Tellez-Plaza, M.; Guallar, E.; De Marco, G.; Lopez-Izquierdo, R.; Gonzalez-Manzano, I.; Carmen Tormos, M.; Martin-Nuñez, G.M.; Rojo-Martinez, G.; Saez, G.T.; et al. Plasma Selenium Levels and Oxidative Stress Biomarkers: A Gene–Environment Interaction Population-Based Study. Free Radic. Biol. Med. 2014, 74, 229–236. [Google Scholar] [CrossRef]
  130. Ojeda, M.L.; Carreras, O.; Nogales, F. The Role of Selenoprotein Tissue Homeostasis in MetS Programming: Energy Balance and Cardiometabolic Implications. Antioxidants 2022, 11, 394. [Google Scholar] [CrossRef]
  131. Kipp, A.P.; Strohm, D.; Brigelius-Flohé, R.; Schomburg, L.; Bechthold, A.; Leschik-Bonnet, E.; Heseker, H. Revised Reference Values for Selenium Intake. J. Trace Elem. Med. Biol. 2015, 32, 195–199. [Google Scholar] [CrossRef]
  132. Juszczak, M.; Bąkowska, M.; Uchwał, P.; Juszczak, M. Content of Selenium in Selected Food Products on the Markets of North-Western Poland. J. Elem. 2018, 24, 111–123. [Google Scholar] [CrossRef]
  133. Pappa, E.C.; Pappas, A.C.; Surai, P.F. Selenium Content in Selected Foods from the Greek Market and Estimation of the Daily Intake. Sci. Total Environ. 2006, 372, 100–108. [Google Scholar] [CrossRef]
  134. Bodnar, M.; Konieczka, P.; Namiesnik, J. The Properties, Functions, and Use of Selenium Compounds in Living Organisms. J. Environ. Sci. Health Part C 2012, 30, 225–252. [Google Scholar] [CrossRef]
  135. Misra, S.; Boylan, M.; Selvam, A.; Spallholz, J.; Björnstedt, M. Redox-Active Selenium Compounds—From Toxicity and Cell Death to Cancer Treatment. Nutrients 2015, 7, 3536–3556. [Google Scholar] [CrossRef]
  136. Vinceti, M.; Crespi, C.M.; Bonvicini, F.; Malagoli, C.; Ferrante, M.; Marmiroli, S.; Stranges, S. The Need for a Reassessment of the Safe Upper Limit of Selenium in Drinking Water. Sci. Total Environ. 2013, 443, 633–642. [Google Scholar] [CrossRef]
  137. Whanger, P.D. Selenocompounds in Plants and Animals and Their Biological Significance. J. Am. Coll. Nutr. 2002, 21, 223–232. [Google Scholar] [CrossRef]
  138. Yang, R.; Liu, Y.; Zhou, Z. Selenium and Selenoproteins, from Structure, Function to Food Resource and Nutrition. Food Sci. Technol. Res. 2017, 23, 363–373. [Google Scholar] [CrossRef]
  139. Alexander, J.; Olsen, A.-K. Selenium—A Scoping Review for Nordic Nutrition Recommendations 2023. Food Nutr. Res. 2023, 67, 10320. [Google Scholar] [CrossRef]
  140. Zhou, F.; Yang, W.; Wang, M.; Miao, Y.; Cui, Z.; Li, Z.; Liang, D. Effects of Selenium Application on Se Content and Speciation in Lentinula Edodes. Food Chem. 2018, 265, 182–188. [Google Scholar] [CrossRef]
  141. Combs, G.F., Jr. Selenium in global food systems. Br. J. Nutr. 2001, 85, 517–547. [Google Scholar] [CrossRef]
  142. Brown, K.; Arthur, J. Selenium, selenoproteins and human health: A review. Public Health Nutr. 2001, 4, 593–599. [Google Scholar] [CrossRef]
  143. Staicu, L.C.; van Hullebusch, E.D.; Oturan, M.A.; Ackerson, C.J.; Lens, P.N. Removal of colloidal biogenic selenium from wastewater. Chemosphere 2015, 125, 130–138. [Google Scholar] [CrossRef] [PubMed]
  144. Mehdi, Y.; Hornick, J.L.; Istasse, L.; Dufrasne, I. Selenium in the environment, metabolism and involvement in body functions. Molecules 2013, 18, 3292–3311. [Google Scholar] [CrossRef] [PubMed]
  145. Gorini, F.; Sabatino, L.; Pingitore, A.; Vassalle, C. Selenium: An Element of Life Essential for Thyroid Function. Molecules 2021, 26, 7084. [Google Scholar] [CrossRef]
  146. Sumana, S.L.; Chen, H.; Shui, Y.; Zhang, C.; Yu, F.; Zhu, J.; Su, S. Effect of Dietary Selenium on the Growth and Immune Systems of Fish. Animals 2023, 13, 2978. [Google Scholar] [CrossRef] [PubMed]
  147. Tan, L.C.; Nancharaiah, Y.V.; van Hullebusch, E.D.; Lens, P.N.L. Selenium: Environmental significance, pollution, and biological treatment technologies. Biotechnol. Adv. 2016, 34, 886–907. [Google Scholar] [CrossRef]
  148. Farooq, M.R.; Zhang, Z.; Yuan, L.; Liu, X.; Rehman, A.; Bañuelos, G.S.; Yin, X. Influencing Factors on Bioavailability and Spatial Distribution of Soil Selenium in Dry Semi-Arid Area. Agriculture 2023, 13, 576. [Google Scholar] [CrossRef]
  149. Kieliszek, M.; Błażejak, S. Current Knowledge on the Importance of Selenium in Food for Living Organisms: A Review. Molecules 2016, 21, 609. [Google Scholar] [CrossRef]
  150. Hadrup, N.; Ravn-Haren, G. Absorption, distribution, metabolism and excretion (ADME) of oral selenium from organic and inorganic sources: A review. J. Trace Elem. Med. Biol. 2021, 67, 126801. [Google Scholar] [CrossRef]
  151. Thiry, C.; Ruttens, A.; De Temmerman, L.; Schneider, Y.-J.; Pussemier, L. Current knowledge in species-related bioavailability of selenium in food. Food Chem. 2012, 130, 767–784. [Google Scholar] [CrossRef]
  152. Hardy, G.; Hardy, I.; Manzanares, W. Selenium supplementation in the critically ill. Nutr. Clin. Pract. 2012, 27, 21–33. [Google Scholar] [CrossRef] [PubMed]
  153. Forceville, X. Effects of high doses of selenium, as sodium selenite, in septic shock patients a placebo-controlled, randomized, double-blind, multi-center phase II study—Selenium and sepsis. J. Trace Elem. Med. Biol. 2007, 21, 62–65. [Google Scholar] [CrossRef] [PubMed]
  154. Vinceti, M.; Wei, E.T.; Malagoli, C.; Bergomi, M.; Vivoli, G. Adverse Health Effects of Selenium in Humans. Rev. Environ. Health 2001, 16, 233. [Google Scholar] [CrossRef] [PubMed]
  155. MacFarquhar, J.K. Acute Selenium Toxicity Associated with a Dietary Supplement. Arch. Intern. Med. 2010, 170, 256. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, G.; Wang, S.; Zhou, R.; Sun, S. Endemic Selenium Intoxication of Humans in China. Am. J. Clin. Nutr. 1983, 37, 872–881. [Google Scholar] [CrossRef]
  157. Raisbeck, M.F. Selenosis. Vet. Clin. N. Am. Food Anim. Pract. 2000, 16, 465–480. [Google Scholar] [CrossRef] [PubMed]
  158. Wei, J.; Zeng, C.; Gong, Q.; Yang, H.; Li, X.; Lei, G.; Yang, T. The Association between Dietary Selenium Intake and Diabetes: A Cross-Sectional Study among Middle-Aged and Older Adults. Nutr. J. 2015, 14, 18. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, Z.; Chang, C.; Zhang, Y.; Chai, Z.; Li, J.; Qiu, C. The Association between Serum Selenium Concentration and Prognosis in Patients with Heart Failure in a Chinese Population. Sci. Rep. 2021, 11, 14533. [Google Scholar] [CrossRef]
  160. Hsueh, Y.-M.; Su, C.-T.; Shiue, H.-S.; Chen, W.-J.; Pu, Y.-S.; Lin, Y.-C.; Tsai, C.-S.; Huang, C.-Y. Levels of Plasma Selenium and Urinary Total Arsenic Interact to Affect the Risk for Prostate Cancer. Food Chem. Toxicol. 2017, 107, 167–175. [Google Scholar] [CrossRef]
  161. Alahmar, A.T. The Effect of Selenium Therapy on Semen Parameters, Antioxidant Capacity, and Sperm DNA Fragmentation in Men with Idiopathic Oligoasthenoteratospermia. Biol. Trace Elem. Res. 2023, 201, 5671–5676. [Google Scholar] [CrossRef] [PubMed]
  162. Chen, M.; Sun, Y.; Wu, Y. Lower Circulating Zinc and Selenium Levels Are Associated with an Increased Risk of Asthma: Evidence from a Meta-Analysis. Public Health Nutr. 2020, 23, 1555–1562. [Google Scholar] [CrossRef] [PubMed]
  163. Shi, Y.; Yang, W.; Tang, X.; Yan, Q.; Cai, X.; Wu, F. Keshan Disease: A Potentially Fatal Endemic Cardiomyopathy in Remote Mountains of China. Front. Pediatr. 2021, 9, 576916. [Google Scholar] [CrossRef] [PubMed]
  164. Institute of Medicine. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids: A Report of the Panel on Dietary Antioxidants and Related Compounds, Subcommittees on Upper Reference Levels of Nutrients and of Interpretation and Use of Dietary Reference Intakes, and the Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Food and Nutrition Board, Institute of Medicine; Institute of Medicine (U.S.), Ed.; National Academy Press: Washington, DC, USA, 2000; ISBN 978-0-309-06949-6. [Google Scholar]
  165. Pei, J.; Fu, W.; Yang, L.; Zhang, Z.; Liu, Y. Oxidative Stress Is Involved in the Pathogenesis of Keshan Disease (an Endemic Dilated Cardiomyopathy) in China. Oxid. Med. Cell. Longev. 2013, 2013, 1–5. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, L.; Feng, H.; Fu, S.; Sun, D. Low Selenium and Low Protein Exacerbate Myocardial Damage in Keshan Disease by Affecting the PINK1/Parkin-Mediated Mitochondrial Autophagy Pathway. Curr. Med. Sci. 2024, 44, 93–101. [Google Scholar] [CrossRef]
  167. Segovia-Cubero, J.; Ruiz-Bautista, L.; Maiz-Carro, L.; Girón-Moreno, R.M.; Prados-Sánchez, M.C.; Martínez-Martínez, M.T.; González-Estecha, M.; Mingo-Santos, S.; Gómez-Bueno, M.; Salas-Antón, C.; et al. The Cardiomyopathy of Cystic Fibrosis: A Modern Form of Keshan Disease. Front. Cardiovasc. Med. 2024, 11, 1285223. [Google Scholar] [CrossRef]
  168. An, P.; Wan, S.; Luo, Y.; Luo, J.; Zhang, X.; Zhou, S.; Xu, T.; He, J.; Mechanick, J.I.; Wu, W.-C.; et al. Micronutrient Supplementation to Reduce Cardiovascular Risk. J. Am. Coll. Cardiol. 2022, 80, 2269–2285. [Google Scholar] [CrossRef]
  169. Zhang, X.; Liu, C.; Guo, J.; Song, Y. Selenium Status and Cardiovascular Diseases: Meta-Analysis of Prospective Observational Studies and Randomized Controlled Trials. Eur. J. Clin. Nutr. 2016, 70, 162–169. [Google Scholar] [CrossRef]
Figure 2. Food products rich in Se. Based on the studies of Juszczak et al. [132] and Pappa et al. [133].
Figure 2. Food products rich in Se. Based on the studies of Juszczak et al. [132] and Pappa et al. [133].
Antioxidants 13 00688 g002
Figure 3. Soil properties affecting the bioavailability of selenium to plants [148].
Figure 3. Soil properties affecting the bioavailability of selenium to plants [148].
Antioxidants 13 00688 g003
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

Leszto, K.; Biskup, L.; Korona, K.; Marcinkowska, W.; Możdżan, M.; Węgiel, A.; Młynarska, E.; Rysz, J.; Franczyk, B. Selenium as a Modulator of Redox Reactions in the Prevention and Treatment of Cardiovascular Diseases. Antioxidants 2024, 13, 688. https://doi.org/10.3390/antiox13060688

AMA Style

Leszto K, Biskup L, Korona K, Marcinkowska W, Możdżan M, Węgiel A, Młynarska E, Rysz J, Franczyk B. Selenium as a Modulator of Redox Reactions in the Prevention and Treatment of Cardiovascular Diseases. Antioxidants. 2024; 13(6):688. https://doi.org/10.3390/antiox13060688

Chicago/Turabian Style

Leszto, Klaudia, Laura Biskup, Klaudia Korona, Weronika Marcinkowska, Maria Możdżan, Andrzej Węgiel, Ewelina Młynarska, Jacek Rysz, and Beata Franczyk. 2024. "Selenium as a Modulator of Redox Reactions in the Prevention and Treatment of Cardiovascular Diseases" Antioxidants 13, no. 6: 688. https://doi.org/10.3390/antiox13060688

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