Next Article in Journal
Selenoprotein K Is Essential for the Migration and Phagocytosis of Immature Dendritic Cells
Next Article in Special Issue
NCOA4 Regulates Iron Recycling and Responds to Hepcidin Activity and Lipopolysaccharide in Macrophages
Previous Article in Journal
Cold Argon Athmospheric Plasma for Biomedicine: Biological Effects, Applications and Possibilities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Associations between Circulating SELENOP Level and Disorders of Glucose and Lipid Metabolism: A Meta-Analysis

1
School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
2
Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
3
Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, USA
4
Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, 119146 Moscow, Russia
5
Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia
6
Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
7
Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou 510080, China
*
Author to whom correspondence should be addressed.
Antioxidants 2022, 11(7), 1263; https://doi.org/10.3390/antiox11071263
Submission received: 9 May 2022 / Revised: 15 June 2022 / Accepted: 23 June 2022 / Published: 27 June 2022
(This article belongs to the Special Issue Trace Elements Metabolism and Oxidative Stress)

Abstract

:
Selenoprotein P (SELENOP) is an extracellular antioxidant, selenium transporter, and hepatokine interfering with glucose and lipid metabolism. To study the association between the circulating SELENOP concentration and glucose and lipid metabolic diseases (GLMDs), including gestational diabetes (GD), metabolic syndrome (MetS), non-alcoholic fatty liver disease, obesity, and type 2 diabetes, as well as the individual markers, a meta-analysis was conducted by searching multiple databases from their establishment through March 2022 and including 27 articles published between October 2010 and May 2021, involving 4033 participants. Participants with GLMDs had higher levels of SELENOP than those without GLMDs (standardized mean difference = 0.84, 95% CI: 0.16 to 1.51), and the SELENOP levels were positively correlated with the markers of GLMDs (pooled effect size = 0.09, 95% CI: 0.02 to 0.15). Subgroup analyses showed that the SELENOP concentrations were higher in women with GD and lower in individuals with MetS than their counterparts, respectively. Moreover, SELENOP was positively correlated with low-density lipoprotein cholesterol, but not with the other markers of GLMDs. Thus, the heterogenicity derived from diseases or disease markers should be carefully considered while interpreting the overall positive association between SELENOP and GLMDs. Studies with a larger sample size and advanced design are warranted to confirm these findings.

Graphical Abstract

1. Introduction

Globally, the prevalence of glucose and lipid metabolic diseases (GLMDs) is higher than that of all other diseases, posing serious threats to human health. The most frequently occurring GLMDs include dyslipidemia, gestational diabetes (GD), metabolic syndrome (MetS), non-alcoholic fatty liver disease (NAFLD) [1], obesity, and type 2 diabetes (T2D). They often coexist and may share a common pathophysiology [2]. Recent studies have shown that the liver, as the “hub” organ of GLMDs [3], can release several secreted proteins called hepatokines to regulate glucose and lipid metabolism (GLM) under stress conditions [4].
Selenoprotein P (SELENOP) is a liver-derived selenium (Se) carrier and a hepatokine in circulation. A single human SELENOP molecule contains up to ten selenocysteines covalently bounding Se. The first selenocysteine near the N-terminal of SELENOP has been demonstrated to have antioxidant properties, while the remaining nine concentrated at its C-terminal are mainly used to transport Se [5,6,7,8,9,10]. Studies have reported the relationship between SELENOP and several components and key indicators of GLMDs [11,12,13,14], but the results were inconsistent [4,15,16,17,18,19,20,21,22,23,24,25,26]. Although several meta-analyses have investigated the association of Se exposure [27,28] or Se supplementation [29,30] with the risk of GLMDs, none of the previous meta-analyses comprehensively examined the details of the association of SELENOP with major GLMDs, their individual components, and key indicators. In this study, we aimed to extend the previous studies by performing a further detailed meta-analysis for the purpose of adding to new evidence for future interventions with drugs and lifestyle factor changes to improve the prevention and control of the diseases.

2. Materials and Methods

2.1. Literature Search

This study was registered with PROSPERO in October 2021 and accepted for inclusion in November 2021 (registration ID number CRD42021257310), and the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) 2020 assertion [31] were strictly followed.
Two qualified investigators independently searched the following databases: PubMed, Embase, Web of Science, The Cochrane Library, China Biology Medicine, WanFang Data, VIP Database, and China National Knowledge Infrastructure, from their establishments to 14 March 2022. The searched keywords were (“selenoprotein P” OR “Selp” OR “Sepp” OR “Sepp1” OR “SELENOP”) AND (“glucose and lipid metabolism” OR “glucolipid metabolic disease” OR “dyslipidemia” OR “fatty liver” OR “obesity” OR “diabetes” OR “atherosclerosis” OR “hypertriglyceridemia” OR “metabolic syndrome” OR “glucose” OR “lipid” OR “insulin resistance” OR “fatty acid” OR “cholesterol” OR “triglycerides” OR “triglyceride” OR “body mass index” OR “BMI”), without restriction to any part of the publications. Furthermore, the references cited within the relevant articles were also reviewed in order to identify additional studies. All of the retrieved articles were published in English or Chinese and managed using the reference manager software EndNote X9 (Clarivate Analytics, Philadelphia, PA, USA).

2.2. Study Identification and Selection

Two independent reviewers performed the study identification and selection. Articles were included if they met all of the following criteria: (1) studies on GLMDs and GLM; and (2) presenting the concentrations of SELENOP in both patients and controls or at least one correlation coefficient between GLM indicators and SELENOP. The exclusion criteria were: (1) duplicate study on the same population; (2) irrelevant study judged from the title and abstract; (3) reviews, case reports, letters, editorials, abstracts, comments, and unpublished articles; (4) study with only animal or cellular experiment(s); (5) lack of predefined outcome data required for analyses; or (6) the evaluation score of the methodological quality (detailed in the next paragraph) being < 6. If there was a disagreement between the two independent researchers (R.Y. and Z.W.) for one study, its eligibility was reevaluated by a third investigator. The corresponding authors of studies with missing data or inaccessible full text were contacted.

2.3. Quality Assessment and Data Extraction

The Newcastle-Ottawa Scale [32,33] was used to assess the methodological quality of the finally included studies, with scores of 7–10 being high quality, 4–6 being moderate quality, and 0–3 being poor quality [34]. Following the inclusion and exclusion criteria, two of the authors assessed and extracted data from the studies independently after reading the key information of the articles, which included: (1) study characteristics, including author(s), year of publication, study country, ethnicity, disease, study design, adjusted factors, and sample size; (2) specimen, concentration, and detection method for SELENOP; and (3) type of correlation (Pearson correlation coefficient (PCC), Spearman correlation coefficient (SCC), or r2) between SELENOP and the GLM indicator. If an article had stratification analyses and reported SELENOP concentrations for different genotypes of a single nucleotide polymorphism (SNP), the reported concentrations were considered as those for different studies in our meta-analysis. However, if multiple correlation coefficient values were reported based on the same relationship between a GLM indicator and SELENOP, the total population or adjusted correlation coefficients were selected for inclusion. Disagreements in the data extraction process were resolved by group discussions between the authors.

2.4. Meta-Analysis

For the SELENOP concentration, data were expressed as the means and standard deviations (SDs); otherwise, the reported standard error (SE), median and interquartile range, or geometric mean and SE were converted to the expression by the corresponding formula [35,36,37,38]. For the correlation between SELENOP and GLM indicators, data were expressed as PCC, and SCC was converted to PCC using an appropriate method [39].
Standardized mean differences (SMDs) and 95% confidence intervals (CIs) were calculated to assess the differences in the SELENOP concentrations between groups. The associations between SELENOP and GLM indicators were evaluated through the combined correlation coefficients (r) (presented as the effect sizes (ESs) in the forest plots) and 95% CIs. The heterogeneity between studies was tested by calculating the Q statistic and the inconsistency index (I2) [40,41]. p < 0.05 or I2 > 50% indicated the presence of heterogeneity [42], and the random effect model was adopted. Otherwise, the fixed-effect model was used. Subgroup analysis was performed to examine possible sources of heterogeneity, and sensitivity analysis was conducted to detect the potential outliers. Publication bias was examined using funnel plots, Egger’s test, and Begg’s test.
Statistical analysis was performed using the STATA 15.0 software package (Stata Corporation, College Station, TX, USA). A two-sided p value of < 0.05 was considered statistically significant.

3. Results

3.1. Study Selection

In the first step of searching for published studies, 1967 articles were selected from the proposed search databases, and 1 [43] from the listed references in 1 of those articles [44]. After careful review, we excluded 607 duplicate articles and 1220 articles that did not meet the inclusion criteria (assessed by their study titles and abstracts). Of the 141 articles, we further excluded 114 articles without detail or eligible data for meta-analysis due to unsearchable full-texts (n = 11), animal or cellular studies (n = 13), reviews (n = 48), and others (n = 42). Finally, 27 articles [15,17,18,19,22,23,24,25,26,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60] were included in the meta-analysis (Figure 1).

3.2. Study Characteristics

The selected articles for review were published between October 2010 and May 2021, covering 36 studies among different populations or those with different genotypes. The general characteristics of these studies are summarized in Table 1, with information on the relationships between the SELENOP level and the concerned indicators included in Supplementary Table S1. Of the studies, SMDs were extracted from 31 studies in the 23 articles, and correlation coefficients were obtained from 14 studies in the 13 articles (Supplementary Figure S1). All of the included studies were of observational design, including 15 case-control studies in 9 articles and 21 cross-sectional studies in 18 articles. The sample sizes across the studies ranged from 21 to 905. The predominant method used to measure the concentrations of SELENOP was the enzyme-linked immunosorbent assay (ELISA). The high-performance liquid chromatography combined with inductively coupled plasma-mass spectrometry (HPLC + ICP-MS) method was adopted in two articles studying T2D, and the sol particle homogeneous immunoassay (SPIA) method was used in two articles studying hyperglycemia and overweightness/obesity, respectively. The SELENOP concentration data among the studies were presented as different statistics, though most were means ± SDs. For eight studies [17,18,19,24,45,52,53,59], the PCCs were calculated based on the SCCs provided in the papers (Supplementary Table S1).
Using the modified Newcastle-Ottawa scale [32,33], Table S2 shows the evaluation of the methodological quality of the selected 27 articles. Of them, 26 studies were considered as “high quality”, and the other one as “medium quality”.

3.3. Meta-Analysis

A total of 4033 participants were involved in the included 27 articles, but in the meta-analysis, the number of participants (time per person) was 4292. In one article, participants were studied for different SNP genotypes [50], and another article provided data before and after follow-up [45].

3.3.1. Relationships between SELENOP Level and GLMDs

The overall data revealed that the participants with GLMDs had significantly higher levels of SELENOP than the controls (SMD = 0.84, 95% CI: 0.16 to 1.51, n = 31). The heterogeneity test showed high heterogeneity among studies (p  <  0.001, I2 = 98.2%; Figure 2). However, the results of the sensitivity analysis indicated that, when each study was removed at a time, the overall random effect did not change significantly and no potential outliers were detected (Supplementary Figure S2). Asymmetry was observed in the funnel plot (Supplementary Figure S3), and statistical asymmetry tests also indicated the existence of publication bias (p = 0.003 for Egger’s test and 0.021 for Begg’s test).
Subgroup analyses based on the types of diseases showed that the level of SELENOP in the patients with GD was higher than that of the healthy controls (SMD = 9.47, 95% CI: 0.90 to 18.04, I2 = 99.2%, n = 3). The levels of SELENOP in the MetS patients were lower than those in the controls (SMD = −0.48, 95% CI: −0.65 to −0.30, I2 = 5.6%, n = 8), while the levels of SELENOP in the patients with other diseases were not different from those in the controls, respectively (Figure 2). However, if the sensitive study on NAFLD conducted by Zhang and Hao [54] (Supplementary Figure S4) was excluded, the combined results showed a negative association between SELENOP and NAFLD (SMD = −0.97, 95% CI: −1.51 to −0.42, I2 = 84.6%, Supplementary Figure S5).
In addition, subgroup analyses based on SELENOP detection methods were performed. The overall results showed that the SELENOP levels detected by the ELISA method in the GLMD patients were higher than those in the controls (SMD = 0.84, 95% CI: 0.13 to 1.55, n = 28) (Supplementary Figure S6). Specifically, for T2D, both ELISA (SMD = 0.27, 95% CI: −1.82 to 2.35, n = 5) and HPLC + ICP-MS (SMD = −1.42, 95% CI: −3.07 to 0.24, n = 2) did not find any significant differences in the SELENOP levels between the patients and controls (Supplementary Figure S7). For obesity, the ELISA found no significance (SMD = 0.38, 95% CI: −0.02 to 0.77, n = 5), but the SPIA indicated higher SELENOP levels in patients than in the controls (SMD = 22.96, 95% CI: 15.80 to 30.13, n = 1) (Supplementary Figure S8).

3.3.2. Correlations between SELENOP and GLM Markers

Figure 3 indicates that SELENOP was positively and significantly correlated with the body mass index (BMI), fasting insulin (FIns), fasting plasma/serum glucose (FPG), hemoglobin A1c (HbA1c), high-density lipoprotein cholesterol (HDL-C), homeostasis model assessment of insulin resistance (HOMA-IR), low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC) [22], and triglyceride (TG) (pooled ES = 0.09, 95% CI: 0.02 to 0.15, n = 75). The inter-study heterogeneity across the studies was significant (I2 = 92.1%, p = 0.000, Figure 3). In the subgroup analysis, a positive and significant correlation was only observed between SELENOP and LDL-C (pooled ES = 0.14, 95% CI: 0.01 to 0.27, n = 5, I2 = 73.7%, p = 0.004), but there were significant correlations observed with any other GLM markers. Sensitivity analyses were performed, and the results remained consistent with the pooled effect.

4. Discussion

The plasma/serum Se includes that from SELENOP, glutathione peroxidase (GPX) 3, albumin-bound fraction, free Se, etc. Containing multiple selenocysteine residues, SELENOP accounts for 48–53% of the blood Se [10,61]. Se deficiency may lead to a decrease in SELENOP expression or premature terminations just before one of the selenocysteine residues in the C-terminal, resulting in SELENOP truncates [9,10]. On the other hand, the SELENOP expression may become saturated at a certain high Se level and resist a further increase in the Se level [9]. Therefore, the SELENOP protein abundance assayed using the predominant methods targeting the N-terminal did not parallel the circulation Se concentration, and SELENOP was not representable by Se in terms of the circulating concentration related to GLMDs [45]. Unfortunately, there were few studies meeting our inclusion criteria presenting both Se and SELENOP data, which prevented us from comparing their differences in association with the outcomes by reliable quantification methods, instead of general discussions. More interestingly, SELENOP is cleavable with two kallikrein cutting sites between the first and the second selenocysteine residues [10]. It has been reported that proteases are activated at inflammatory sites [62], and that Se modulates the inflammatory and immune responses [63]. Therefore, it is possible that plasma-kallikrein-processed SELENOP fragments regulate inflammation by controlling Se action in cells, though the cleaved form is about 1.2% of the total SELENOP in the human plasma [64].
Studies have shown that a decrease in SELENOP may cause various dysfunctions related to Se deficiency and oxidative stress [65,66,67,68]. Furthermore, low SELENOP concentrations were strongly associated with the risk of incident cardiovascular disease and mortality from all causes, cardiovascular disease, and COVID-19 [12,69]. Nevertheless, excessive SELENOP may lead to insulin resistance [70], and treatment with the full-length form of SELENOP may impair insulin signal transduction in cultured hepatocytes [46]. In animal experiments, an excess of circulating SELENOP induces both impaired insulin signaling in the peripheral tissues and decreased insulin secretion in the pancreas [71]. Moreover, SELENOP may induce insulin resistance by affecting adipose tissue to reduce the adiponectin levels [72] or by acting on cultured myotubes with low-density lipoprotein (LDL) receptor-associated protein 1 [73], thereby leading to diabetes. On the other hand, insulin exerts inhibitory effects on the gene expression of SELENOP in hepatocytes [46,74,75], and impaired insulin action in certain metabolic disorders might increase the expression and circulating level of SELENOP. Therefore, the overproduction of SELENOP and the development of metabolic disorders might reinforce one another mutually in a vicious cycle, and SELENOP is a multifunctional protein in the pathology of GLMDs [9,76].

4.1. GD and T2D

During a normal pregnancy, an increase in insulin resistance occurs simultaneously with an increase in oxidative stress, which is particularly prominent in women with GD [77]. Growing attention has been paid to the association between SELENOP and GD [78,79,80,81,82,83,84]. Two recent meta-analyses on Se and GD observed that the serum Se levels in GD patients were significantly lower than those in the healthy pregnancy group [85,86]. However, the results of our meta-analysis showed that the SELENOP concentrations were elevated in patients with GD compared with normal pregnant women, which is consistent with a recently published meta-analysis on the relationship between hepatokines (including only one article for SELENOP) and GD [87]. Though one of the three included articles in our meta-analysis tested both the circulating Se and SELENOP concentrations [15], it seems that there were no obvious clues to understand the relationship between Se and SELENOP in their associations with GD by indicating no difference in the Se levels (76–78 µg/L), but SELENOP in GD women was lower compared with their counterparts.
One study showed that the SELENOP concentration at baseline in the oral glucose test was associated with the future post-load plasma glucose in male participants, whereas, in female participants, it was interrelated with the future FPG [45]. This suggests that sexual dimorphism is present in the main target organs of SELENOP in humans, and it may help to interpret the positive correlation of SELENOP with GD characterized by upregulated hepatic glucose production.
The overall results of the nine T2D studies included in our meta-analysis [18,19,43,44,56] showed that the SELENOP concentrations in T2D were similar to those without T2D, which was inconsistent with the results of the previous meta-analysis studies on the relationship between Se and T2D [27,28,88,89], suggesting disparity between Se and SELENOP in their associations with diseases. In previous studies, the investigators suggested that there was a U-shaped relationship between Se and T2D. Either a significantly decreased or elevated Se level, lower or higher than the normal physiological range, should be considered as a risk factor for T2D [90,91,92,93]. Of the two included studies for our meta-analysis that simultaneously measured the concentrations of Se and SELENOP [43,44], both Se and SELENOP did not differ between the patients and controls when the mean plasma Se was about 80 µg/L [43], but both were significantly higher in patients than in the controls when the mean serum Se was > 94 µg/L [44]. However, in another recently published cohort study that did not meet our inclusion criteria, the serum Se (with median value at 80 µg/L), but not SELENOP, was associated with increased risk of developing T2D in the final adjustment model [94]. Thus, the circulating Se levels may affect the correlation between SELENOP and T2D, but this needs to be further verified by more studies.

4.2. MetS and NAFLD

MetS is a complex disease defined by a set of interrelated metabolic factors [95]. The combined results of the articles included in this study showed that lower SELENOP levels were associated with increased risk of MetS, which is consistent with the findings of a recently published review [96]. Though the only article included in our meta-analysis on MetS presenting both Se and SELENOP data suggested no difference in the serum Se and lower SELENOP in patients than in controls [50], another meta-analysis on the association between the dietary Se level and MetS indicated their negative association [97].
Two articles that were not included in our study due to data skewness also showed lower SELENOP concentrations in MetS patients [52,98]. Patients with MetS are usually in an inflammation state, coupled with impaired liver functions [99]. Thus, the circulating SELENOP, as a negative acute-phase reactant and a hepatokine, could be downregulated by both inflammation [51,100,101,102] and decreased selenoprotein synthetic capacity in the MetS status.
NAFLD is considered to be the hepatic symptom of MetS due to its coexistence with visceral obesity, insulin resistance, and dyslipidemia [1], and its primary characteristic is the accumulation of lipids in the liver accompanied by lipid peroxidation, oxidative stress, inflammation, etc. [103]. Though SELENOP has been shown to play an important role in the pathological process of lipid accumulation [104], the relationship between SELENOP and NAFLD remains unclear. Our sensitivity analysis on the NAFLD subgroup of GLMDs indicated that the study conducted by Zhang and Hao [54] may affect the stability of the null association between SELELOP and NAFLD, though it did meet our inclusion criteria. If the study was excluded, the lower SELENOP levels in NAFLD patients were consistent with the relationship between MetS and SELENOP. However, the SELENOP concentrations were higher in pregnant women with NAFLD and increased the risk of GD [16], which suggested that some specific physiological conditions may complicate the association. Other confounding variables may also add to the heterogeneity between studies and affect their results. Furthermore, the higher SELENOP levels in NAFLD in some studies may imply a protective mechanism that counteracts the higher oxidative stress in the initial stage of diseases, but the mechanism may be insufficient in the advanced stages, such as definite non-alcoholic steatohepatitis [24], cirrhosis [105], and hepatocellular carcinoma [106], in which lower SELENOP levels have been reported.

4.3. Obesity and BMI

Oxidative stress in obesity leads to metabolic and endocrine dysfunction of adipose tissue, contributing to the development of obesity-related insulin resistance [107,108]. SELENOP has been shown to respond significantly to this proinflammatory stimulus [96]. A previous review pointed out that the circulating SELENOP levels were elevated in patients with obesity [96], but our meta-analysis of the included studies found no difference in the SELENOP levels between patients with obesity and healthy subjects, further confirming a recent meta-analysis on the Se (n = 14 for children and 18 for adults) and SELENOP (n = 3, which were among our included 6 studies) levels in people with overweightness and obesity [109]. The most likely reason is that the individuals with obesity were not able to be separated from the overweight and obese individuals, because almost all of the included studies defined the cases with BMI > 25 kg/m2 for adults. If subjects with obesity defined by a higher cut-off value of BMI were compared with normal-weight subjects, a statistically significant relationship between obesity and the SELENOP levels might be observed. In addition, two of the included studies recruited individuals with anemia [47] or NAFLD [25], which might also have had a certain impact on the results.
BMI is commonly used to define overweightness and obesity in clinical settings and in epidemiological studies [110]. As a measure of relative weight, it is directly linked to health risks and mortality in many populations [111]. Both BMI [112] and SELENOP are associated with insulin resistance and inflammation, and the relationship between BMI and SELENOP is still controversial. The results of our meta-analysis showed that SELENOP was not statistically correlated with BMI, but the heterogeneity was high. The possible reasons are as follows: the included subjects were not all obese, and the distributions of age, race, and gender of each study population varied, which might affect the levels of SELENOP and BMI, as well as their relationship [45,113].

4.4. Lipid Profiles

Meta-analyses have shown that Se supplementation does not affect the lipid levels or only results in a statistically significant improvement in the TC, TG, and/or VLDL-C levels [29,30]. A similar kind of inconsistency also existed in the association between SELENOP and the lipid profiles. Some evidence showed that SELENOP was positively correlated with HDL-C [17,21,48,52], LDL-C [26,58], TC [22,52], and TG [18,21,55] in the plasma/serum. In contrast, some cross-sectional studies reported that higher SELENOP levels were correlated with lower HDL-C [55], TC [48,58], and TG [48,52,58], while others indicated no correlation with LDL-C [18,52]. Given the paradox, we conducted a comprehensive meta-analysis of the relationship between SELENOP and lipid profiles. The results showed that SELENOP was positively correlated with LDL-C, but not with other lipid indices.
LDL, carrying cholesterol in the form of LDL-C, circulates in the plasma and supplies various cells with cholesterol under normal physiological conditions, but oxidized LDL has cytotoxic effects and is thought to be involved in the development of atherosclerosis, a type of GLMD [114]. Having the ability to reduce phospholipid hydroperoxides and bind to glycosaminoglycans [115], SELENOP may play an antioxidant protective role by binding to ApoB-100 [116], a glycosylated LDL component, and SELENOP can protect LDL against oxidation in a cell-free in vitro system [114]. In response to the oxidized LDL potentially existing in diseases with high LDL-C levels included in our study, SELENOP might be elevated. In addition, studies have suggested that the role of SELENOP in GD and T2D might reduce the adiponectin levels by affecting adipose tissue [20,72], and the adiponectin levels are known to be negatively correlated with LDL-C [117]. This can also account for the positive relationship between SELENOP and LDL-C.

4.5. Glucose Metabolism

Se and SELENOP are closely related to glucose metabolism. For Se, a recent study showed a linear relationship between Se deficiency and hypoglycemia in healthy adults. The mechanism is suggested to be the low Se status potentially causing diminished activity of GPX1 (one of the selenoproteins) in insulin target cells, contributing to amplified insulin signals due to the dysregulation of redox-regulated proteins, such as the insulin-antagonistic protein tyrosine phosphatase 1B (PTP1B). In contrast, an elevated Se concentration upregulates PTP1B and induces suppressed insulin signaling and hyperglycemia [118]. A certain concentration of serum Se is essential to maintain the expression of GPX or SELENOP, which seems to be required for achieving and maintaining euglycemia. The relationship between SELENOP and serum glucose in the Se-deficient state needs further study. Furthermore, a high glucose concentration stimulates the pancreas to secrete insulin and the liver to release SELENOP [53], while excessive SELENOP worsens glucose metabolism via insulin resistance and the impairment of insulin secretion [18,46,71]. Therefore, the causal relationship between an increased SELENOP concentration and metabolic disorders of glucose has not been clarified [119]. Moreover, the correlations between SELENOP and glucose metabolism indicators (FIns, FPG, HbA1c, and HOMA-IR) have not been uniformly established yet.
Elevations in FPG and HbA1c reflected acute dysregulated glucose metabolism and chronic hyperglycemia, respectively [120], and HOMA-IR, another glucose metabolism indicator, is based on insulin resistance and hyperinsulinemia [121]. A previous cross-sectional study showed that moderate positive correlations were observed between the SELENOP levels and FPG and HbA1c among T2D patients [20], and the hepatic mRNA abundance of SELENOP was also positively correlated with FPG [46]. In addition, a Korean study reported that serum SELENOP was positively correlated with HOMA-IR in NAFLD patients [21]. In contrast, more recent epidemiologic findings showed inverse relationships between the SELENOP levels and FIns, FPG, HbA1c, HOMA-IR, and several other metabolic traits in adults [49,52] and young children [48]. However, a follow-up study found no significant associations between the baseline serum SELENOP concentrations and these metabolic markers [45]. We pooled these inconsistent results and found that SELENOP was not associated with indicators of glucose metabolism in the population, which suggested that the concentrations of SELENOP, FPG, and HbA1c for participants of a healthy condition and with various diseases, as well as their different stages, may not achieve a consistent correlation.

4.6. SNPs of SELENOP

The SNPs of the SELENOP gene were found to be correlated with certain metabolic phenotypes. In a meta-analysis that included three different ethnic groups, rs28919926 and rs146125471 showed associations with acute insulin resistance, and rs7579 with the insulin sensitivity index [122]. The rs7579 A allele is associated with a decrease in the SELENOP levels in subjects with or without MetS, and MetS decreases the SELENOP levels in general, except for the rs7579 AA homozygote carriers. However, rs3877899 AA coordinates with MetS to decrease the SELENOP levels [50]. In Turkish pregnant women, the rs13154178 GG genotype was coupled with a higher SELENOP concentration in GD patients [15], and the G allele was positively associated with FPG and GD occurrence [72]. Another study in pregnant women from the United Kingdom showed that, under Se supplementation during pregnancy, the rs3877899 A allele helped to maintain the Se status at a constant level, though the activity of GPX3, another circulating selenoprotein, increased [123]. Moreover, SELENOP rs3877899 altered the LDL levels in response to Brazil nut intake, suggesting that SELENOP polymorphisms affected the ability of Se to improve lipid biomarkers [124]. In general, very few studies have considered variations in SELENOP in its association with GLMDs, which needs more attention in the future.

4.7. Limitations and Advantages

Several limitations of our study should be kept in mind while interpreting the results. First, the studies used different laboratory methods to test the SELENOP concentrations. For example, results from ELISA, HPLC + ICP-MS, and SPIA may lead to high heterogeneity in the synthesis results. Second, potential publication bias may have occurred. Several reviewed studies in the report had a small sample size, and several studies applied a cross-sectional study design, which is unable to test any causality. Third, we could not obtain the original datasets of the reviewed studies, which made it impossible to use the ab initio analysis of data to conduct analysis in detail, such as to test a dose-response relationship. Fourth, unobserved bias may occur due to the included studies with different confounding factors adjusted.
Despite the limitations discussed above, this study has two important strengths. First, the study focusing on the relationships of SELENOP with several GLMDs and related indicators is novel. Thus, building upon the recently published studies, our meta-analysis study with a robust design provides updated evidence on SELENOP studies in the body of literature. Second, we used SMD and correlation-coefficient-derived ES to evaluate the relationships and examined multiple factors. These analysis approaches enhanced the statistical power and led to the results of our study being more explanatory.

5. Conclusions

In conclusion, among the major GLMDs, there was a positive correlation between increased circulating SELENOP concentration and the risk of GD and elevated LDL-C concentration, but a negative correlation with MetS. Further epidemiological studies with a larger sample size, advanced study designs, and especially comparison of Se and SELENOP in their associations with GLMDs are needed to test the specific causal association between SELENOP and GLMDs and/or SELENOP’s value for the prediction and treatment of GLMDs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox11071263/s1, Figure S1: Articles reporting the concentrations and/or correlation coefficients of selenoprotein P; Figure S2: Sensitivity analysis of the association between selenoprotein P and metabolic disorders of glucose and lipids; Figure S3: Funnel plot of the meta-analysis of the association between selenoprotein P and metabolic disorders of glucose and lipids; Figure S4: Sensitivity analysis of the selenoprotein P level in non-alcoholic fatty liver disease or healthy control patients; Figure S5: Forest plot of the selenoprotein P level in non-alcoholic fatty liver disease or healthy control patients after the exclusion of one study; Figure S6: Subgroup analysis based on the selenoprotein P detection method for total participants; Figure S7: Subgroup analysis based on the selenoprotein P detection method for T2D; Figure S8: Subgroup analysis based on the selenoprotein P detection method for obesity; Table S1: Characteristics of the included studies (with indicators and correlation coefficients supplementing Table 1); Table S2: The methodological quality evaluation of the included articles using the modified Newcastle-Ottawa scale. References [125,126] are cited in the supplementary materials.

Author Contributions

Conceptualization, R.Y. and J.-C.Z.; Methodology, R.Y., Z.W. and J.-C.Z.; Validation, M.M., P.X. and J.-C.Z.; Formal analysis, R.Y. and Z.W.; Investigation, R.Y., Z.W. and P.X.; Resources, J.-C.Z.; Data curation, R.Y., Z.W., M.M. and J.-C.Z.; Writing—original draft, R.Y., Z.W. and P.X.; Writing—review and editing, R.Y., Z.W., M.M., L.L., A.A.T., X.G.L. and J.-C.Z.; Visualization, P.X. and J.-C.Z.; Supervision, J.-C.Z.; Project administration, J.-C.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Science, Technology and Innovation Commission of Shenzhen Municipality, China (grant number JCYJ20200109142446804) and the National Natural Science Foundation of China (grant number 81973038).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article and Supplementary Material.

Acknowledgments

The authors acknowledge the anonymous reviewers for their helpful comments and constructive suggestions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lazarus, J.V.; Mark, H.E.; Anstee, Q.M.; Arab, J.P.; Batterham, R.L.; Castera, L.; Cortez-Pinto, H.; Crespo, J.; Cusi, K.; Dirac, M.A.; et al. Advancing the global public health agenda for NAFLD: A consensus statement. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 60–78. [Google Scholar] [CrossRef] [PubMed]
  2. Hoffman, D.J.; Powell, T.L.; Barrett, E.S.; Hardy, D.B. Developmental origins of metabolic diseases. Physiol. Rev. 2021, 101, 739–795. [Google Scholar] [CrossRef] [PubMed]
  3. Jones, J.G. Hepatic glucose and lipid metabolism. Diabetologia 2016, 59, 1098–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Misu, H. Identification of hepatokines involved in pathology of type 2 diabetes and obesity. Endocr. J. 2019, 66, 659–662. [Google Scholar] [CrossRef] [Green Version]
  5. Saito, Y.; Hayashi, T.; Tanaka, A.; Watanabe, Y.; Suzuki, M.; Saito, E.; Takahashi, K. Selenoprotein P in human plasma as an extracellular phospholipid hydroperoxide glutathione peroxidase—Isolation and enzymatic characterization of human selenoprotein P. J. Biol. Chem. 1999, 274, 2866–2871. [Google Scholar] [CrossRef] [Green Version]
  6. Takebe, G.; Yarimizu, J.; Saito, Y.; Hayashi, T.; Nakamura, H.; Yodoi, J.; Nagasawa, S.; Takahashi, K. A comparative study on the hydroperoxide and thiol specificity of the glutathione peroxidase family and selenoprotein P. J. Biol. Chem. 2002, 277, 41254–41258. [Google Scholar] [CrossRef] [Green Version]
  7. Burk, R.F.; Hill, K.E. Selenoprotein P-expression, f.functions, and roles in mammals. Biochim. Biophys. Acta 2009, 1790, 1441–1447. [Google Scholar] [CrossRef] [Green Version]
  8. Kurokawa, S.; Eriksson, S.; Rose, K.L.; Wu, S.; Motley, A.K.; Hill, S.; Winfrey, V.P.; McDonald, W.H.; Capecchi, M.R.; Atkins, J.F.; et al. Sepp1(UF) forms are N-terminal selenoprotein P truncations that have peroxidase activity when coupled with thioredoxin reductase-1. Free Radic. Biol. Med. 2014, 69, 67–76. [Google Scholar] [CrossRef] [Green Version]
  9. Burk, R.F.; Hill, K.E. Regulation of selenium metabolism and transport. Annu. Rev. Nutr. 2015, 35, 109–134. [Google Scholar] [CrossRef]
  10. Saito, Y. Selenium transport mechanism via selenoprotein P-its physiological role and related diseases. Front. Nutr. 2021, 8, 685517. [Google Scholar] [CrossRef]
  11. Fernandez-Ruiz, I. Selenoprotein P—A new player in PAH. Nat. Rev. Cardiol. 2018, 15, 381. [Google Scholar] [CrossRef] [PubMed]
  12. 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] [PubMed] [Green Version]
  13. Saito, Y. Selenoprotein P as a significant regulator of pancreatic beta cell function. J. Biochem. 2020, 167, 119–124. [Google Scholar] [CrossRef] [PubMed]
  14. Polyzos, S.A.; Kountouras, J.; Goulas, A.; Duntas, L. Selenium and selenoprotein P in nonalcoholic fatty liver disease. Hormones 2020, 19, 61–72. [Google Scholar] [CrossRef] [PubMed]
  15. Cinemre, F.B.S.; Cinemre, H.; Yucel, A.; Degirmencioglu, S.; Tuten, A.; Yuksel, M.A.; Yilmaz, N.; Gulyasar, T.; Yildiz, M.; Bahtiyar, N.; et al. The role of selenoprotein P and selenium in the etiopathogenesis of gestational diabetes mellitus: Association with selenoprotein P1 gene (rs3877899) polymorphism. Trace Elem. Electroly. 2018, 35, 174–182. [Google Scholar] [CrossRef]
  16. Lee, S.M.; Kwak, S.H.; Koo, J.N.; Oh, I.H.; Kwon, J.E.; Kim, B.J.; Kim, S.M.; Kim, S.Y.; Kim, G.M.; Joo, S.K.; et al. Non-alcoholic fatty liver disease in the first trimester and subsequent development of gestational diabetes mellitus. Diabetologia 2019, 62, 238–248. [Google Scholar] [CrossRef] [Green Version]
  17. Altinova, A.E.; Iyidir, O.T.; Ozkan, C.; Ors, D.; Ozturk, M.; Gulbahar, O.; Bozkurt, N.; Toruner, F.B.; Akturk, M.; Cakir, N.; et al. Selenoprotein P is not elevated in gestational diabetes mellitus. Gynecol. Endocrinol. 2015, 31, 874–876. [Google Scholar] [CrossRef]
  18. Yang, S.J.; Hwang, S.Y.; Choi, H.Y.; Yoo, H.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Baik, S.H.; Choi, D.S.; Choi, K.M. Serum selenoprotein P levels in patients with type 2 diabetes and prediabetes: Implications for insulin resistance, inflammation, and atherosclerosis. J. Clin. Endocrinol. Metab. 2011, 96, E1325–E1329. [Google Scholar] [CrossRef]
  19. Roman, M.; Lapolla, A.; Jitaru, P.; Sechi, A.; Cosma, C.; Cozzi, G.; Cescon, P.; Barbante, C. Plasma selenoproteins concentrations in type 2 diabetes mellitus—A pilot study. Transl. Res. 2010, 156, 242–250. [Google Scholar] [CrossRef]
  20. Misu, H.; Ishikura, K.; Kurita, S.; Takeshita, Y.; Ota, T.; Saito, Y.; Takahashi, K.; Kaneko, S.; Takamura, T. Inverse correlation between serum levels of selenoprotein P and adiponectin in patients with type 2 diabetes. PLoS ONE 2012, 7, e34952. [Google Scholar] [CrossRef] [Green Version]
  21. Choi, H.Y.; Hwang, S.Y.; Lee, C.H.; Hong, H.C.; Yang, S.J.; Yoo, H.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Baik, S.H.; et al. Increased selenoprotein P levels in subjects with visceral obesity and nonalcoholic fatty liver disease. Diabetes Metab. J. 2013, 37, 63–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chen, M.; Liu, B.; Wilkinson, D.; Hutchison, A.T.; Thompson, C.H.; Wittert, G.A.; Heilbronn, L.K. Selenoprotein P is elevated in individuals with obesity, but is not independently associated with insulin resistance. Obes. Res. Clin. Pract. 2017, 11, 227–232. [Google Scholar] [CrossRef] [PubMed]
  23. Sargeant, J.A.; Aithal, G.P.; Takamura, T.; Misu, H.; Takayama, H.; Douglas, J.A.; Turner, M.C.; Stensel, D.J.; Nimmo, M.A.; Webb, D.R.; et al. The influence of adiposity and acute exercise on circulating hepatokines in normal-weight and overweight/obese men. Appl. Physiol. Nutr. Metab. 2018, 43, 482–490. [Google Scholar] [CrossRef] [Green Version]
  24. Polyzos, S.A.; Kountouras, J.; Mavrouli, M.; Katsinelos, P.; Doulberis, M.; Gavana, E.; Duntas, L. Selenoprotein P in patients with nonalcoholic fatty liver disease. Exp. Clin. Endocrinol. Diabetes 2019, 127, 598–602. [Google Scholar] [CrossRef] [PubMed]
  25. Flisiak-Jackiewicz, M.; Bobrus-Chociej, A.; Wasilewska, N.; Tarasow, E.; Wojtkowska, M.; Lebensztejn, D.M. Can hepatokines be regarded as novel non-invasive serum biomarkers of intrahepatic lipid content in obese children? Adv. Med. Sci. 2019, 64, 280–284. [Google Scholar] [CrossRef]
  26. Cetindagli, I.; Kara, M.; Tanoglu, A.; Ozalper, V.; Aribal, S.; Hancerli, Y.; Unal, M.; Ozari, O.; Hira, S.; Kaplan, M.; et al. Evaluation of endothelial dysfunction in patients with nonalcoholic fatty liver disease: Association of selenoprotein P with carotid intima-media thickness and endothelium-dependent vasodilation. Clin. Res. Hepatol. Gastroenterol. 2017, 41, 516–524. [Google Scholar] [CrossRef]
  27. Vinceti, M.; Filippini, T.; Wise, L.A.; Rothman, K.J. A systematic review and dose-response meta-analysis of exposure to environmental selenium and the risk of type 2 diabetes in nonexperimental studies. Environ. Res. 2021, 197, 111210. [Google Scholar] [CrossRef]
  28. Vinceti, M.; Filippini, T.; Rothman, K.J. Selenium exposure and the risk of type 2 diabetes: A systematic review and meta-analysis. Eur. J. Epidemiol. 2018, 33, 789–810. [Google Scholar] [CrossRef]
  29. Hasani, M.; Djalalinia, S.; Sharifi, F.; Varmaghani, M.; Zarei, M.; Abdar, M.E.; Asayesh, H.; Noroozi, M.; Kasaeian, A.; Gorabi, A.M.; et al. Effect of selenium supplementation on lipid profile: A systematic review and meta-analysis. Horm. Metab. Res. 2018, 50, 715–727. [Google Scholar] [CrossRef]
  30. Tabrizi, R.; Akbari, M.; Moosazadeh, M.; Lankarani, K.B.; Heydari, S.T.; Kolahdooz, F.; Mohammadi, A.A.; Shabani, A.; Badehnoosh, B.; Jamilian, M.; et al. The effects of selenium supplementation on glucose metabolism and lipid profiles among patients with metabolic diseases: A systematic review and meta-analysis of randomized controlled trials. Horm. Metab. Res. 2017, 49, 826–830. [Google Scholar] [CrossRef]
  31. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  32. Herzog, R.; Alvarez-Pasquin, M.J.; Diaz, C.; Del Barrio, J.L.; Estrada, J.M.; Gil, A. Are healthcare workers’ intentions to vaccinate related to their knowledge, beliefs and attitudes? A systematic review. BMC Public Health 2013, 13, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Stang, A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur. J. Epidemiol. 2010, 25, 603–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pitukweerakul, S.; Thavaraputta, S.; Prachuapthunyachart, S.; Karnchanasorn, R. Hypovitaminosis D is associated with psoriasis: A systematic review and meta-analysis. Kans. J. Med. 2019, 12, 103–108. [Google Scholar] [CrossRef]
  35. Higgins, J.P.T.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A. (Eds.) Cochrane Handbook for Systematic Reviews of Interventions, 2nd ed.; John Wiley & Sons: Chichester, UK, 2019. [Google Scholar]
  36. Luo, D.; Wan, X.; Liu, J.; Tong, T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Stat. Methods Med. Res. 2018, 27, 1785–1805. [Google Scholar] [CrossRef] [Green Version]
  37. Wan, X.; Wang, W.; Liu, J.; Tong, T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med. Res. Methodol. 2014, 14, 135. [Google Scholar] [CrossRef] [Green Version]
  38. Shi, J.; Luo, D.; Wan, X.; Liu, Y.; Liu, J.; Bian, Z.; Tong, T. Detecting the skewness of data from the sample size and the five-number summary. arXiv 2020, arXiv:2010.05749. [Google Scholar]
  39. Rupinski, M.T.; Dunlap, W.P. Approximating A basic introduction to fixed-effect. Educ. Psychol. Meas. 1996, 56, 419–429. [Google Scholar] [CrossRef]
  40. Leeflang, M.M.; Deeks, J.J.; Gatsonis, C.; Bossuyt, P.M.; Cochrane Diagnostic Test Accuracy Working, G. Systematic reviews of diagnostic test accuracy. Ann. Intern. Med. 2008, 149, 889–897. [Google Scholar] [CrossRef]
  41. Zamora, J.; Abraira, V.; Muriel, A.; Khan, K.; Coomarasamy, A. Meta-DiSc: A software for meta-analysis of test accuracy data. BMC Med. Res. Methodol. 2006, 6, 31. [Google Scholar] [CrossRef]
  42. Higgins, J.P.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gonzalez de Vega, R.; Fernandez-Sanchez, M.L.; Fernandez, J.C.; Alvarez Menendez, F.V.; Sanz-Medel, A. Selenium levels and glutathione peroxidase activity in the plasma of patients with type II diabetes mellitus. J. Trace Elem. Med. Biol. 2016, 37, 44–49. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Q.; Li, W.; Wang, J.; Hu, B.; Yun, H.; Guo, R.; Wang, L. Selenium levels in community dwellers with type 2 diabetes mellitus. Biol. Trace Elem. Res. 2019, 191, 354–362. [Google Scholar] [CrossRef]
  45. Oo, S.M.; Misu, H.; Saito, Y.; Tanaka, M.; Kato, S.; Kita, Y.; Takayama, H.; Takeshita, Y.; Kanamori, T.; Nagano, T.; et al. Serum selenoprotein P, but not selenium, predicts future hyperglycemia in a general Japanese population. Sci. Rep. 2018, 8, 16727. [Google Scholar] [CrossRef] [PubMed]
  46. Misu, H.; Takamura, T.; Takayama, H.; Hayashi, H.; Matsuzawa-Nagata, N.; Kurita, S.; Ishikura, K.; Ando, H.; Takeshita, Y.; Ota, T.; et al. A liver-derived secretory protein, selenoprotein P, causes insulin resistance. Cell Metab. 2010, 12, 483–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Larvie, D.Y.; Doherty, J.L.; Donati, G.L.; Armah, S.M. Relationship between selenium and hematological markers in young adults with normal weight or overweight/obesity. Antioxidants 2019, 8, 463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Ko, B.J.; Kim, S.M.; Park, K.H.; Park, H.S.; Mantzoros, C.S. Levels of circulating selenoprotein P, fibroblast growth factor (FGF) 21 and FGF23 in relation to the metabolic syndrome in young children. Int. J. Obes. 2014, 38, 1497–1502. [Google Scholar] [CrossRef]
  49. Gharipour, M.; Sadeghi, M.; Salehi, M.; Behmanesh, M.; Khosravi, E.; Dianatkhah, M.; Haghjoo Javanmard, S.; Razavi, R.; Gharipour, A. Association of expression of selenoprotein P in mRNA and protein levels with metabolic syndrome in subjects with cardiovascular disease: Results of the Selenegene study. J. Gene Med. 2017, 19, e2945. [Google Scholar] [CrossRef]
  50. Gharipour, M.; Ouguerram, K.; Nazih, E.H.; Salehi, M.; Behmanesh, M.; Razavi, R.; Gharipour, A.; Diantkhah, M.; Sadeghi, M. Effect of single nucleotide polymorphisms in SEPS1 and SEPP1 on expression in the protein level in metabolic syndrome in subjects with cardiovascular disease. Mol. Biol. Rep. 2019, 46, 5685–5693. [Google Scholar] [CrossRef]
  51. El-Kafrawy, N.A.F.; Atta, A.E.B.M.; Abdelsattar, S.; Zewain, S.K.E.D. Serum selenoprotein P in lean and obese Egyptian individuals and its relation to insulin resistance. Alex. J. Med. 2021, 57, 61–69. [Google Scholar] [CrossRef]
  52. di Giuseppe, R.; Koch, M.; Schlesinger, S.; Borggrefe, J.; Both, M.; Muller, H.P.; Kassubek, J.; Jacobs, G.; Nothlings, U.; Lieb, W. Circulating selenoprotein P levels in relation to MRI-derived body fat volumes, liver fat content, and metabolic disorders. Obesity 2017, 25, 1128–1135. [Google Scholar] [CrossRef] [PubMed]
  53. Caviglia, G.P.; Rosso, C.; Armandi, A.; Gaggini, M.; Carli, F.; Abate, M.L.; Olivero, A.; Ribaldone, D.G.; Saracco, G.M.; Gastaldelli, A.; et al. Interplay between oxidative stress and metabolic derangements in non-alcoholic fatty liver disease: The role of selenoprotein P. Int. J. Mol. Sci. 2020, 21, 8838. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, L.; Hao, Y. Study on selenoprotein P in type 2 diabetes mellitus and non-alcoholic fatty liver disease. J. Baotou Med. 2018, 34, 95–96. [Google Scholar] [CrossRef]
  55. Pan, J.; Yu, H.; Lei, Z.; Han, J.; Jia, W. Selenoprotein P in type 2 diabetes mellitus and its association with insulin resistance. Natl. Med. J. Chin. 2014, 94, 1710–1713. [Google Scholar] [CrossRef]
  56. Jin, S.; Jin, M.; Zhang, Y. Correlation between serum selenoprotein P and renal injury and prognosis in patients with diabetic nephropathy. J. Trop. Med. 2020, 20, 1589–1592. [Google Scholar] [CrossRef]
  57. Jiang, L.; Dou, X.; Chen, Y. Clinical analysis of serum selenoprotein P in patients with gestational diabetes mellitus. Chin. Fore. Med. Res. 2019, 17, 7–9. [Google Scholar] [CrossRef]
  58. Fan, H.; Shi, B.; Li, H.; Sun, H. Correlation analysis between serum selenoprotein P and non -alcoholic fatty liver disease in elders with type 2 diabetes mellitus. Chin. J. Diabetes Mellit. 2019, 27, 250–253. [Google Scholar] [CrossRef]
  59. Chen, Y.; He, X.J.; Chen, X.Y.; Li, Y.M.; Ke, Y.N. SeP is elevated in NAFLD and participates in NAFLD pathogenesis through AMPK/ACC pathway. J. Cell Physiol. 2021, 236, 3800–3807. [Google Scholar] [CrossRef]
  60. Jung, T.W.; Ahn, S.H.; Shin, J.W.; Kim, H.C.; Park, E.S.; Abd El-Aty, A.M.; Hacimuftuoglu, A.; Song, K.H.; Jeong, J.H. Protectin DX ameliorates palmitate-induced hepatic insulin resistance through AMPK/SIRT1-mediated modulation of fetuin-A and SeP expression. Clin. Exp. Pharmacol. Physiol. 2019, 46, 898–909. [Google Scholar] [CrossRef]
  61. Zheng, W.Y.; He, R.; Boada, R.; Subirana, M.A.; Ginman, T.; Ottosson, H.; Valiente, M.; Zhao, Y.; Hassan, M. A general covalent binding model between cytotoxic selenocompounds and albumin revealed by mass spectrometry and X-ray absorption spectroscopy. Sci. Rep. 2020, 10, 1274. [Google Scholar] [CrossRef] [Green Version]
  62. Esmon, C.T.; Fukudome, K.; Mather, T.; Bode, W.; Regan, L.M.; Stearns-Kurosawa, D.J.; Kurosawa, S. Inflammation, sepsis, and coagulation. Haematologica 1999, 84, 254–259. [Google Scholar] [PubMed]
  63. Peretz, A.M.; Neve, J.D.; Famaey, J.P. Selenium in rheumatic diseases. Semin. Arthritis Rheum. 1991, 20, 305–316. [Google Scholar] [CrossRef]
  64. Saito, Y.; Sato, N.; Hirashima, M.; Takebe, G.; Nagasawa, S.; Takahashi, K. Domain structure of bi-functional selenoprotein P. Biochem. J. 2004, 381, 841–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Saito, Y.; Yoshida, Y.; Akazawa, T.; Takahashi, K.; Niki, E. Cell death caused by selenium deficiency and protective effect of antioxidants. J. Biol. Chem. 2003, 278, 39428–39434. [Google Scholar] [CrossRef] [Green Version]
  66. Hill, K.E.; Zhou, J.; McMahan, W.J.; Motley, A.K.; Atkins, J.F.; Gesteland, R.F.; Burk, R.F. Deletion of selenoprotein P alters distribution of selenium in the mouse. J. Biol. Chem. 2003, 278, 13640–13646. [Google Scholar] [CrossRef] [Green Version]
  67. Saito, Y.; Shichiri, M.; Hamajima, T.; Ishida, N.; Mita, Y.; Nakao, S.; Hagihara, Y.; Yoshida, Y.; Takahashi, K.; Niki, E.; et al. Enhancement of lipid peroxidation and its amelioration by vitamin E in a subject with mutations in the SBP2 gene. J. Lipid Res. 2015, 56, 2172–2182. [Google Scholar] [CrossRef] [Green Version]
  68. Suzuki, T.; Kelly, V.P.; Motohashi, H.; Nakajima, O.; Takahashi, S.; Nishimura, S.; Yamamoto, M. Deletion of the selenocysteine tRNA gene in macrophages and liver results in compensatory gene induction of cytoprotective enzymes by Nrf2. J. Biol. Chem. 2008, 283, 2021–2030. [Google Scholar] [CrossRef] [Green Version]
  69. Moghaddam, A.; Heller, R.A.; Sun, Q.; Seelig, J.; Cherkezov, A.; Seibert, L.; Hackler, J.; Seemann, P.; Diegmann, J.; Pilz, M.; et al. Selenium deficiency is associated with mortality risk from COVID-19. Nutrients 2020, 12, 2098. [Google Scholar] [CrossRef]
  70. Takamura, T. Hepatokine selenoprotein P-mediated reductive stress causes resistance to intracellular signal transduction. Antioxid. Redox Signal. 2020, 33, 517–524. [Google Scholar] [CrossRef]
  71. Mita, Y.; Nakayama, K.; Inari, S.; Nishito, Y.; Yoshioka, Y.; Sakai, N.; Sotani, K.; Nagamura, T.; Kuzuhara, Y.; Inagaki, K.; et al. Selenoprotein P-neutralizing antibodies improve insulin secretion and glucose sensitivity in type 2 diabetes mouse models. Nat. Commun. 2017, 8, 1658. [Google Scholar] [CrossRef]
  72. Akbaba, G.; Akbaba, E.; Sahin, C.; Kara, M. The relationship between gestational diabetes mellitus and selenoprotein-P plasma 1 (SEPP1) gene polymorphisms. Gynecol. Endocrinol. 2018, 34, 849–852. [Google Scholar] [CrossRef] [PubMed]
  73. Misu, H.; Takayama, H.; Saito, Y.; Mita, Y.; Kikuchi, A.; Ishii, K.; Chikamoto, K.; Kanamori, T.; Tajima, N.; Lan, F.; et al. Deficiency of the hepatokine selenoprotein P increases responsiveness to exercise in mice through upregulation of reactive oxygen species and AMP-activated protein kinase in muscle. Nat. Med. 2017, 23, 508–516. [Google Scholar] [CrossRef] [PubMed]
  74. Walter, P.L.; Steinbrenner, H.; Barthel, A.; Klotz, L.O. Stimulation of selenoprotein P promoter activity in hepatoma cells by FoxO1a transcription factor. Biochem. Biophys. Res. Commun. 2008, 365, 316–321. [Google Scholar] [CrossRef] [PubMed]
  75. Speckmann, B.; Walter, P.L.; Alili, L.; Reinehr, R.; Sies, H.; Klotz, L.O.; Steinbrenner, H. Selenoprotein P expression is controlled through interaction of the coactivator PGC-1alpha with FoxO1a and hepatocyte nuclear factor 4alpha transcription factors. Hepatology 2008, 48, 1998–2006. [Google Scholar] [CrossRef]
  76. Chen, J.; Berry, M.J. Selenium and selenoproteins in the brain and brain diseases. J. Neurochem. 2003, 86, 1–12. [Google Scholar] [CrossRef]
  77. Chen, X.; Scholl, T.O.; Leskiw, M.J.; Donaldson, M.R.; Stein, T.P. Association of glutathione peroxidase activity with insulin resistance and dietary fat intake during normal pregnancy. J. Clin. Endocrinol. Metab. 2003, 88, 5963–5968. [Google Scholar] [CrossRef] [Green Version]
  78. Tan, M.; Sheng, L.; Qian, Y.; Ge, Y.; Wang, Y.; Zhang, H.; Jiang, M.; Zhang, G. Changes of serum selenium in pregnant women with gestational diabetes mellitus. Biol. Trace Elem. Res. 2001, 83, 231–237. [Google Scholar] [CrossRef]
  79. Al-Saleh, E.; Nandakumaran, M.; Al-Shammari, M.; Al-Harouny, A. Maternal-fetal status of copper, iron, molybdenum, selenium and zinc in patients with gestational diabetes. J. Matern. Fetal Neonatal. Med. 2004, 16, 15–21. [Google Scholar] [CrossRef]
  80. Bo, S.; Lezo, A.; Menato, G.; Gallo, M.L.; Bardelli, C.; Signorile, A.; Berutti, C.; Massobrio, M.; Pagano, G.F. Gestational hyperglycemia, zinc, selenium, and antioxidant vitamins. Nutrition 2005, 21, 186–191. [Google Scholar] [CrossRef]
  81. Kilinc, M.; Guven, M.A.; Ezer, M.; Ertas, I.E.; Coskun, A. Evaluation of serum selenium levels in Turkish women with gestational diabetes mellitus, glucose intolerants, and normal controls. Biol. Trace Elem. Res. 2008, 123, 35–40. [Google Scholar] [CrossRef]
  82. Al-Saleh, E.; Nandakumaran, M.; Al-Rashdan, I.; Al-Harmi, J.; Al-Shammari, M. Maternal-foetal status of copper, iron, molybdenum, selenium and zinc in obese gestational diabetic pregnancies. Acta Diabetol. 2007, 44, 106–113. [Google Scholar] [CrossRef] [PubMed]
  83. Hamdan, H.Z.; Elbashir, L.M.; Hamdan, S.Z.; Elhassan, E.M.; Adam, I. Zinc and selenium levels in women with gestational diabetes mellitus at Medani Hospital, Sudan. J. Obstet. Gynaecol. 2014, 34, 567–570. [Google Scholar] [CrossRef] [PubMed]
  84. Molnar, J.; Garamvolgyi, Z.; Herold, M.; Adanyi, N.; Somogyi, A.; Rigo, J., Jr. Serum selenium concentrations correlate significantly with inflammatory biomarker high-sensitive CRP levels in Hungarian gestational diabetic and healthy pregnant women at mid-pregnancy. Biol. Trace Elem. Res. 2008, 121, 16–22. [Google Scholar] [CrossRef] [PubMed]
  85. Askari, G.; Iraj, B.; Salehi-Abargouei, A.; Fallah, A.A.; Jafari, T. The association between serum selenium and gestational diabetes mellitus: A systematic review and meta-analysis. J. Trace Elem. Med. Biol. 2015, 29, 195–201. [Google Scholar] [CrossRef] [PubMed]
  86. Kong, F.J.; Ma, L.L.; Chen, S.P.; Li, G.; Zhou, J.Q. Serum selenium level and gestational diabetes mellitus: A systematic review and meta-analysis. Nutr. J. 2016, 15, 94. [Google Scholar] [CrossRef] [Green Version]
  87. Cai, Z.; Yang, Y.; Zhang, J. Hepatokine levels during the first or early second trimester of pregnancy and the subsequent risk of gestational diabetes mellitus: A systematic review and meta-analysis. Biomarkers 2021, 26, 517–531. [Google Scholar] [CrossRef]
  88. Kohler, L.N.; Foote, J.; Kelley, C.P.; Florea, A.; Shelly, C.; Chow, H.S.; Hsu, P.; Batai, K.; Ellis, N.; Saboda, K.; et al. Selenium and type 2 diabetes: Systematic review. Nutrients 2018, 10, 1924. [Google Scholar] [CrossRef] [Green Version]
  89. Kim, J.; Chung, H.S.; Choi, M.K.; Roh, Y.K.; Yoo, H.J.; Park, J.H.; Kim, D.S.; Yu, J.M.; Moon, S. Association between serum selenium level and the presence of diabetes mellitus: A meta-analysis of observational studies. Diabetes Metab. J. 2019, 43, 447–460. [Google Scholar] [CrossRef]
  90. Wang, X.L.; Yang, T.B.; Wei, J.; Lei, G.H.; Zeng, C. Association between serum selenium level and type 2 diabetes mellitus: A non-linear dose-response meta-analysis of observational studies. Nutr. J. 2016, 15, 48. [Google Scholar] [CrossRef] [Green Version]
  91. Rayman, M.P. Selenium and human health. Lancet 2012, 379, 1256–1268. [Google Scholar] [CrossRef]
  92. Rayman, M.P.; Stranges, S. Epidemiology of selenium and type 2 diabetes: Can we make sense of it? Free Radic. Biol. Med. 2013, 65, 1557–1564. [Google Scholar] [CrossRef] [PubMed]
  93. Rocourt, C.R.; Cheng, W.H. Selenium supranutrition: Are the potential benefits of chemoprevention outweighed by the promotion of diabetes and insulin resistance? Nutrients 2013, 5, 1349–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Cabral, M.; Kuxhaus, O.; Eichelmann, F.; Kopp, J.F.; Alker, W.; Hackler, J.; Kipp, A.P.; Schwerdtle, T.; Haase, H.; Schomburg, L.; et al. Trace element profile and incidence of type 2 diabetes, cardiovascular disease and colorectal cancer: Results from the EPIC-Potsdam cohort study. Eur. J. Nutr. 2021, 60, 3267–3278. [Google Scholar] [CrossRef] [PubMed]
  95. Eckel, R.H.; Alberti, K.G.; Grundy, S.M.; Zimmet, P.Z. The metabolic syndrome. Lancet 2010, 375, 181–183. [Google Scholar] [CrossRef]
  96. Tinkov, A.A.; Ajsuvakova, O.P.; Filippini, T.; Zhou, J.C.; Lei, X.G.; Gatiatulina, E.R.; Michalke, B.; Skalnaya, M.G.; Vinceti, M.; Aschner, M.; et al. Selenium and selenoproteins in adipose tissue physiology and obesity. Biomolecules 2020, 10, 658. [Google Scholar] [CrossRef]
  97. Ding, J.; Liu, Q.; Liu, Z.; Guo, H.; Liang, J.; Zhang, Y. Associations of the dietary iron, copper, and selenium level with metabolic syndrome: A meta-analysis of observational studies. Front. Nutr. 2021, 8, 810494. [Google Scholar] [CrossRef]
  98. di Giuseppe, R.; Plachta-Danielzik, S.; Koch, M.; Nöthlings, U.; Schlesinger, S.; Borggrefe, J.; Both, M.; Müller, H.P.; Kassubek, J.; Jacobs, G.; et al. Dietary pattern associated with selenoprotein P and MRI-derived body fat volumes, liver signal intensity, and metabolic disorders. Eur. J. Nutr. 2019, 58, 1067–1079. [Google Scholar] [CrossRef]
  99. Wang, S.; Zhang, J.; Zhu, L.; Song, L.L.; Meng, Z.W.; Jia, Q.; Li, X.; Liu, N.; Hu, T.P.; Zhou, P.P.; et al. Association between liver function and metabolic syndrome in Chinese men and women. Sci. Rep. 2017, 7, 44844. [Google Scholar] [CrossRef]
  100. Renko, K.; Hofmann, P.J.; Stoedter, M.; Hollenbach, B.; Behrends, T.; Kohrle, J.; Schweizer, U.; Schomburg, L. Down-regulation of the hepatic selenoprotein biosynthesis machinery impairs selenium metabolism during the acute phase response in mice. FASEB J. 2009, 23, 1758–1765. [Google Scholar] [CrossRef]
  101. Hesse-Bahr, K.; Dreher, I.; Kohrle, J. The influence of the cytokines Il-1beta and INFgamma on the expression of selenoproteins in the human hepatocarcinoma cell line HepG2. Biofactors 2000, 11, 83–85. [Google Scholar] [CrossRef]
  102. Dallmeier, D.; Larson, M.G.; Vasan, R.S.; Keaney, J.F.; Fontes, J.D.; Meigs, J.B.; Fox, C.S.; Benjamin, E.J. Metabolic syndrome and inflammatory biomarkers: A community-based cross-sectional study at the Framingham Heart Study. Diabetol. Metab. Syndr. 2012, 4, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Day, C.P.; James, O.F. Steatohepatitis: A tale of two “hits”? Gastroenterology 1998, 114, 842–845. [Google Scholar] [CrossRef]
  104. Gu, L.; Zhang, Y.; Zhang, S.; Zhao, H.; Wang, Y.; Kan, D.; Zhang, Y.; Guo, L.; Lv, J.; Hao, Q.; et al. Coix lacryma-jobi seed oil reduces fat accumulation in nonalcoholic fatty liver disease by inhibiting the activation of the p-AMPK/SePP1/ApoER2 pathway. J. Oleo Sci. 2021, 70, 685–696. [Google Scholar] [CrossRef] [PubMed]
  105. Burk, R.F.; Hill, K.E.; Motley, A.K.; Byrne, D.W.; Norsworthy, B.K. Selenium deficiency occurs in some patients with moderate-to-severe cirrhosis and can be corrected by administration of selenate but not selenomethionine: A randomized controlled trial. Am. J. Clin. Nutr. 2015, 102, 1126–1133. [Google Scholar] [CrossRef] [Green Version]
  106. Grgurevic, I.; Podrug, K.; Mikolasevic, I.; Kukla, M.; Madir, A.; Tsochatzis, E.A. Natural history of nonalcoholic fatty liver disease: Implications for clinical practice and an individualized approach. Can. J. Gastroenterol. Hepatol. 2020, 2020, 9181368. [Google Scholar] [CrossRef]
  107. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
  108. Rudich, A.; Kanety, H.; Bashan, N. Adipose stress-sensing kinases: Linking obesity to malfunction. Trends Endocrinol. Metab. 2007, 18, 291–299. [Google Scholar] [CrossRef]
  109. Fontenelle, L.C.; Cardoso de Araújo, D.S.; da Cunha Soares, T.; Clímaco Cruz, K.J.; Henriques, G.S.; Marreiro, D.D.N. Nutritional status of selenium in overweight and obesity: A systematic review and meta-analysis. Clin. Nutr. 2022, 41, 862–884. [Google Scholar] [CrossRef]
  110. Chooi, Y.C.; Ding, C.; Magkos, F. The epidemiology of obesity. Metabolism 2019, 92, 6–10. [Google Scholar] [CrossRef] [Green Version]
  111. Consultation, W.H.O.E. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet 2004, 363, 157–163. [Google Scholar] [CrossRef]
  112. Zierle-Ghosh, A.; Jan, A. Physiology, Body Mass Index. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  113. Letsiou, S.; Nomikos, T.; Panagiotakos, D.B.; Pergantis, S.A.; Fragopoulou, E.; Pitsavos, C.; Stefanadis, C.; Antonopoulou, S. Gender-specific distribution of selenium to serum selenoproteins: Associations with total selenium levels, age, smoking, body mass index, and physical activity. Biofactors 2014, 40, 524–535. [Google Scholar] [CrossRef] [PubMed]
  114. Traulsen, H.; Steinbrenner, H.; Buchczyk, D.P.; Klotz, L.O.; Sies, H. Selenoprotein P protects low-density lipoprotein against oxidation. Free Radic. Res. 2004, 38, 123–128. [Google Scholar] [CrossRef] [PubMed]
  115. Arteel, G.E.; Franken, S.; Kappler, J.; Sies, H. Binding of selenoprotein P to heparin: Characterization with surface plasmon resonance. Biol. Chem. 2000, 381, 265–268. [Google Scholar] [CrossRef] [PubMed]
  116. Esterbauer, H.; Gebicki, J.; Puhl, H.; Jurgens, G. The role of lipid peroxidation and antioxidants in oxidative modification of LDL. Free Radic. Biol. Med. 1992, 13, 341–390. [Google Scholar] [CrossRef]
  117. Matsuzawa, Y. Adiponectin: Identification, physiology and clinical relevance in metabolic and vascular disease. Atheroscler. Suppl. 2005, 6, 7–14. [Google Scholar] [CrossRef]
  118. Wang, Y.; Rijntjes, E.; Wu, Q.; Lv, H.; Gao, C.; Shi, B.; Schomburg, L. Selenium deficiency is linearly associated with hypoglycemia in healthy adults. Redox Biol. 2020, 37, 101709. [Google Scholar] [CrossRef]
  119. Mao, J.; Teng, W. The relationship between selenoprotein P and glucose metabolism in experimental studies. Nutrients 2013, 5, 1937–1948. [Google Scholar] [CrossRef] [Green Version]
  120. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2010, 33 (Suppl. 1), S62–S69. [Google Scholar] [CrossRef] [Green Version]
  121. Matthews, D.R.; Hosker, J.P.; Rudenski, A.S.; Naylor, B.A.; Treacher, D.F.; Turner, R.C. Homeostasis model assessment: Insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 1985, 28, 412–419. [Google Scholar] [CrossRef] [Green Version]
  122. Hellwege, J.N.; Palmer, N.D.; Ziegler, J.T.; Langefeld, C.D.; Lorenzo, C.; Norris, J.M.; Takamura, T.; Bowden, D.W. Genetic variants in selenoprotein P plasma 1 gene (SEPP1) are associated with fasting insulin and first phase insulin response in Hispanics. Gene 2014, 534, 33–39. [Google Scholar] [CrossRef] [Green Version]
  123. Mao, J.Y.; Vanderlelie, J.J.; Perkins, A.V.; Redman, C.W.G.; Ahmadi, K.R.; Rayman, M.P. Genetic polymorphisms that affect selenium status and response to selenium supplementation in United Kingdom pregnant women. Am. J. Clin. Nutr. 2016, 103, 100–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Moriguchi Watanabe, L.; Bueno, A.C.; de Lima, L.F.; Ferraz-Bannitz, R.; Dessordi, R.; Guimaraes, M.P.; Foss-Freitas, M.C.; Barbosa, F.; Navarro, A.M. Genetically determined variations of selenoprotein P are associated with antioxidant, muscular, and lipid biomarkers in response to Brazil nut consumption by patients using statins. Br. J. Nutr. 2022, 127, 679–686. [Google Scholar] [CrossRef] [PubMed]
  125. Borenstein, M.; Hedges, L.V.; Higgins, J.P.; Rothstein, H.R. A basic introduction to fixed-effect and random-effects models for meta-analysis. Res. Synth. Methods 2010, 1, 97–111. [Google Scholar] [CrossRef] [PubMed]
  126. Tsiligianni, I.; Kocks, J.; Tzanakis, N.; Siafakas, N.; van der Molen, T. Factors that influence disease-specific quality of life or health status in patients with COPD: A systematic review and meta-analysis of Pearson correlations. Prim. Care Resp. J. 2011, 20, 257–268. [Google Scholar] [CrossRef]
Figure 1. Strategy for literature search and selection.
Figure 1. Strategy for literature search and selection.
Antioxidants 11 01263 g001
Figure 2. Correlations between the circulating selenoprotein P level and disorders of glucose and lipid metabolism. Abbreviations: GD, gestational diabetes; MetS, metabolic syndrome; NAFLD, non-alcoholic fatty liver disease; SMD, standardized mean difference; T2D, type 2 diabetes; wt, weight.
Figure 2. Correlations between the circulating selenoprotein P level and disorders of glucose and lipid metabolism. Abbreviations: GD, gestational diabetes; MetS, metabolic syndrome; NAFLD, non-alcoholic fatty liver disease; SMD, standardized mean difference; T2D, type 2 diabetes; wt, weight.
Antioxidants 11 01263 g002
Figure 3. Correlations between selenoprotein P and 9 GLM markers. Abbreviations: ES, effect size; FIns, fasting insulin; FPG, fasting plasma/serum glucose; GLM, glucose and lipid metabolism; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TC, total cholesterol; TG, triglyceride; wt, weight.
Figure 3. Correlations between selenoprotein P and 9 GLM markers. Abbreviations: ES, effect size; FIns, fasting insulin; FPG, fasting plasma/serum glucose; GLM, glucose and lipid metabolism; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TC, total cholesterol; TG, triglyceride; wt, weight.
Antioxidants 11 01263 g003
Table 1. Characteristics of the selected studies for meta-analysis.
Table 1. Characteristics of the selected studies for meta-analysis.
Study aCountryDiseasenSample (Unit) Detection Method bLevel c
CaseControlCaseControl
* Altinova et al., 2015 [17]TurkeyGD3035Plasma (ng/mL)ELISA 2 6.2 (4.5–8.2) 7.9 (4.5–10.7)
* Caviglia et al., 2020 [53]ItalyNAFLD57Serum (ng/mL)ELISA 1T3: 11.8
# Cetindağlı et al., 2017 [26]TurkeyNAFLD9337Plasma (ng/mL)ELISA 91574.2 ± 972.1 232.7 ± 371.05
* Chen et al., 2017 [22]AustraliaOW/OB3429Plasma (μg/mL)ELISA 152.3 ± 39.1 14.5 ± 12.8
# Chen et al., 2021 [59]ChinaNAFLD7979Serum (μg/mL)ELISA 113.4 ± 7.0 11.1 ± 7.1
# Cinemre et al., 2018 [15] TurkeyGD8690Plasma (ng/mL)ELISA 835.29 ± 3.00 46.98 ± 4.59
* di Giuseppe et al., 2017 [52]GermanyMetSQ1: 225; Q2: 227; Q3: 228; Q4: 225Serum (mg/mL)ELISA 2 Q1: 2.86 (1.96–3.70) ;
Q2: 4.52 (3.87–5.98) ;
Q3: 6.05 (5.3–28.47) ;
Q4: 11.72 (8.07–15.79)
* El-Kafrawy et al., 2021 [51] EgyptOW/OB5040Serum (mg/L)ELISA 716.18 ± 3.99 4.25 ± 4.27
* Fan et al., 2019 [58]ChinaT2D and NAFLDT2D and NAFLD: 79;
T2D: 61
Serum (ng/mL)ELISA 1T2D and NAFLD: 1341.11 ± 290.51 ;
T2D: 755.77 ± 184.90
* Flisiak-Jackiewicz et al., 2019 [25]PolandNAFLD
Obesity
34
86
52
24
Serum (pg/mL)ELISA 1 19449.5 (13327–28058)
21421 (11566–28058)
21629 (10369.5–27976)
5411 (1618–15135)
* Gharipour et al., 2017 [49]IranMetS6571Serum (ng/mL)ELISA 341.8 ± 6.57 81.5 ± 15.2
# Gharipour et al., 2019 [50]IranMetSrs7579 GG: 2930Serum (ng/mL)ELISA 355.52 ± 16.78 109.48 ± 29.78
rs7579 GA:182236.65 ± 7.41 59.80 ± 22.06
rs7579 AA: 8529.45 ± 1.97 26.65 ± 2.51
rs3877899 GG: 404440.37 ± 8.44 83.91 ± 21.33
rs3877899 GA: 151356.92 ± 23.34 86.42 ± 40.99
rs3877899 AA: 2329.70 ± 4.1 81.95 ± 107.03
# Gonzalez de Vega et al., 2016 [43]SpainT2D7824Plasma (ppb)HPLC + ICP-MS41.9 ± 12.6 50.5 ± 19.1
# Jiang et al., 2019 [57]ChinaGD3030Serum (mmol/L)ELISA 14.85 ± 1.02 2.43 ± 1.04
# Jin et al., 2020 [56]ChinaDN100100Serum (ng/mL)ELISA 1673.18 ± 86.94 973.84 ± 132.27
* Jung et al., 2019 [60]KoreaOW/OB3535Serum (μg/mL)ELISA 22.3 ± 0.1 1.5 ± 0.1
* Ko et al., 2014 [48]KoreaMetS94116Serum (ng/mL)ELISA 2 16.7 ± 2.2 28.6 ± 2.0
* Larvie et al., 2019 [47]AmericaOW/OB3227Plasma (ng/mL)ELISA 4 352.13 (276, 446) 360.77 (290, 450)
* Misu et al., 2010 [46]JapanT2D129Serum (μg/mL)ELISA 96.7 ± 0.9 5.1 ± 1.7
* Oo et al., 2018 [45]JapanHG76Serum (μg/mL)SPIABaseline: 2.51 ± 0.52
* Pan et al., 2014 [55]ChinaT2D15664Serum (mmol/L)ELISA 13.77 ± 1.79 2.34 ± 2.30
* Polyzos et al., 2019 [24]GreeceNAFLD3127Serum (mg/L)ELISA 5SS: 4.2 ± 0.3 ; Borderline NASH: 4.1 ± 0.4 ; Definite NASH: 3.0 ± 0.5 5 ± 0.2
* Roman et al., 2010 [19]ItalyT2D4015Plasma (ng/mL)HPLC + ICP-MS58 ± 9 56 ± 8
* Sargeant et al., 2017 [23]BritainOW/OB
1111Plasma (μg/mL)SPIA2.81 ± 0.30 3.01 ± 0.39
* Yang et al., 2011 [18]KoreaT2D
PreD
40
40
20Serum (ng/mL)ELISA 1 1032.4 (495.9–2149.4) ; 867.3 (516.3–1582.7) 62.0 (252.5–694.5)
# Zhang and Hao, 2018 [54]ChinaT2D
NAFLD
100
100
100
100
Serum (mmol/L)ELISA 63.05 ± 1.20
4.42 ± 1.80
2.33 ± 2.30
2.33 ± 2.30
# Zhang et al., 2019 [44]ChinaT2D176142Serum (ng/mL)ELISA 11811.1 ± 36.3 1688.2 ± 40.5
Note: DN, diabetic nephropathy; ELISA, enzyme-linked immunosorbent assay; GD, gestational diabetes; HG, hyperglycemia; HPLC, high-performance liquid chromatography; ICP-MS, inductively coupled plasma-mass spectrometry; MetS, metabolic syndrome; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; OW/OB, overweight and obesity; PreD, prediabetes; SPIA, sol particle homogeneous immunoassay; SS: simple steatosis; T2D, type 2 diabetes; n, sample size number. a *, cross-sectional study; #, case-control study. b ELISA kits were provided by 1, Cloud-Clone Corp. Houston, TX, USA; 2, Cusabio, Wuhan, China; 3, Eastbiopharm, Hangzhou, China; 4, MyBioSource (San Diego, CA, USA); 5, selenOmed GmbH, Berlin, Germany; 6, Shanghai Runyu Biotechnology Co., Ltd., Shanghai, China; 7, Shanghai Sunred Biological Technology Co., Ltd., Shanghai, China; 8, Shanghai YeHua Biological Technology Co., Ltd. Gical Technology Co., Ltd., Shanghai, China; 9, unknown. c Data were expressed as quartiles (Q1/2/3/4), tertiles (T1/2/3), medians (interquartile ranges) (), means ± SDs (), means ± SEs (), or geometric means ± SDs () for all subjects or patients vs controls.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yu, R.; Wang, Z.; Ma, M.; Xu, P.; Liu, L.; Tinkov, A.A.; Lei, X.G.; Zhou, J.-C. Associations between Circulating SELENOP Level and Disorders of Glucose and Lipid Metabolism: A Meta-Analysis. Antioxidants 2022, 11, 1263. https://doi.org/10.3390/antiox11071263

AMA Style

Yu R, Wang Z, Ma M, Xu P, Liu L, Tinkov AA, Lei XG, Zhou J-C. Associations between Circulating SELENOP Level and Disorders of Glucose and Lipid Metabolism: A Meta-Analysis. Antioxidants. 2022; 11(7):1263. https://doi.org/10.3390/antiox11071263

Chicago/Turabian Style

Yu, Ruirui, Zhoutian Wang, Miaomiao Ma, Ping Xu, Longjian Liu, Alexey A. Tinkov, Xin Gen Lei, and Ji-Chang Zhou. 2022. "Associations between Circulating SELENOP Level and Disorders of Glucose and Lipid Metabolism: A Meta-Analysis" Antioxidants 11, no. 7: 1263. https://doi.org/10.3390/antiox11071263

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