Serum elementomic analysis indicates a panel of elements related with age | Jia | Aging Pathobiology and Therapeutics

Open Access | Research
This work is licensed under a Creative Commons Attribution-ShareAlike 4.0 International License.

Serum elementomic analysis indicates a panel of elements related with age


Kai-Yue Jiaa,#, Hui-Xian Suna,#, Yan-Ru Lia,#, Can Zhaoa, Xiang Lua,*, Wei Gaob,*

a Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
b Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.

# These authors contributed equally to this work.
* Corresponding author: Wei Gao, M.D., Ph.D.
Mailing address: Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao, Nanjing, Jiangsu Province, 210009, China.
Email: drweig1984@outlook.com
* Corresponding author: Xiang Lu, M.D., Ph.D.
Mailing address: Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, No.109 Longmian Avenue, Nanjing, Jiangsu Province, 211166, China.
Email: luxiang66@njmu.edu.cn

Received: 13 December 2023 / Revised: 21 December 2023 / Accepted: 10 January 2024 / Published: 28 March 2024

DOI: 10.31491/APT.2024.03.134

Abstract

Background: Elementomics, which includes metallic and non-metallic elements, is an emerging and promising research field for human diseases. Researchers are focusing on discovering the relationship between elements and various diseases; however, the changes in element concentrations during the process of aging remain unclear.

Materials and Methods: We performed elementomic analysis in the serum of 70 subjects aged 30 to 96 years using inductively coupled plasma mass spectrometry. The subjects were divided into 7 groups with an age range of 10 years. Random forest was used to estimate the variable importance of elements. Linear regression model and restricted cubic spline analysis were performed to screen for elements individually associated with age. Candidate elements were combined by corresponding multivariate linear regression coefficients to generate a risk score representing their collective effect on age.

Results: Among the 62 detected elements, lithium, boron, calcium, titanium, and selenium were identified as the most important predictors of age. There was an increase in lithium and boron as well as a decrease in calcium and titanium with increasing age. The concentration of selenium was elevated before the age of 60, but decreased thereafter. A formula of element risk score was constructed using the respective coefficients from a multivariate linear regression model for the above five elements. The formula = 4.5522 × lithium + 6.0575 × boron - 4.9990 × calcium - 7.0403 × titanium - 0.8849 × selenium.

Conclusion: Elementomics could be a novel and promising non-invasive biomarker for the assessment of senescence.

Keywords

Elementomics, serum, age, inductively coupled plasma mass spectrometry

Introduction

Aging is an inevitable natural phenomenon characterized by a gradual, time-dependent decline in normal physiological functions [1]. It is predicted that by 2050, the elderly population in China will experience a significant surge, reaching up to 400 million individuals aged over 65 years and 150 million individuals aged over 80 years [2, 3]. Biomarkers of aging are quantifiable parameters that reflect both physiological and pathological aging [4]. To better understand the mechanisms of aging, recent studies have focused on the search for novel biomarkers of aging. In addition to the identified aging-related substances and metabolites, such as total homocysteine in blood and 8-dihydroguanosine in urine [5, 6], there has also been a growing interest in investigating the effects of elements in the progression of aging.
Elementomics is the study of metals, metalloids and their relationships with genomes and proteomes [7]. Metal ions play an important role in regulating protein expression in cells, which is essential for maintaining cellular homeostasis and facilitating detoxification processes [8]. Additionally, a substantial number of protein enzymes rely on metal elements to facilitate their catalytic activities [9]. Current perspectives on elements and disease are mainly concerned with cancer, Alzheimer's disease, Parkinson's disease, cardiovascular disease, etc. [10]. Patients with Huntington's disease have been found to have elevated circulating levels of iron, chromium, selenium, zinc, and arsenic, as well as decreased levels of antimony, lead, and vanadium [11]. Zinc levels are decreased and cadmium levels are increased in patients with prostate cancer [12-14]. In the cardiovascular system, iron and calcium deficiencies have been identified to play a critical role in the development of heart failure [15]. Previous studies have found that the levels of several elements are increased in the aorta, blood, and other tissues of aged rats compared with young rats [16-18]. Meanwhile, age-related differences in elements have also been observed in the elderly population [19, 20]. However, few studies have investigated the relationship between elementomics and the process of aging. The use of inductively coupled plasma mass spectrometry (ICP-MS) allows the rapid and accurate measurement of more than 60 elements simultaneously [21, 22]. In the present study, we aimed to explore the changes in blood element concentrations in different age groups.

Materials and methods

Study population

The study population consisted of 70 subjects, aged 30 to 96 years, recruited from the Health Examination Center of Sir Run Run Hospital, Nanjing Medical University. The subjects were divided into 7 groups as a 10-year age range, with 5 males and 5 females in each group. Participants with the following conditions were excluded: (1) acute cardiopulmonary insufficiency; (2) severe renal insufficiency (creatinine clearance rate < 60 mL/min) or severe liver damage (transaminase levels more than twice the normal values); (3) malignant tumors; (4) gastrointestinal diseases; (5) acute or chronic inflammatory diseases; (6) eating disorders or malnutrition; (7) taking any kind of dietary supplements or trace elements before enrollment or during the study; (8) occupationally exposed to pollution or living in a polluted environment. The study was conducted in accordance with the tenets of the Declaration of Helsinki and was approved by the Ethics Committee of Sir Run Run Hospital, Nanjing Medical University (approval number 2019-SR-S041). Written informed consent was obtained from each participant.

Laboratory measurements

For hematologic and biochemical parameters, peripheral venous blood samples were collected after at least 8 hours of fasting. Blood samples were centrifuged at 3000 g for 10 minutes to separate the blood into serum and cell fractions within 2 hours. Serum samples were collected and stored at -80°C prior to further analysis. Fasting blood glucose (FBG), alanine transaminase (ALT), aspartate aminotransferase (AST), total bilirubin (TBil), serum creatinine (SCr), blood urea nitrogen (BUN), total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDLC), albumin (ALB), and globulin (GLOB) were measured using a chemistry analyzer (Olympus AU5400, Chemical Ltd., Japan).

Exposure assessment

Samples were thawed at 4℃ for approximately 1 hour, and then 100 μL of serum was diluted to 1.6 mL with 1% (v/v) HNO3, internal standards (including 40 ppb lithium, 20 ppb rhodium, 20 ppb indium, and 20 ppb rhenium), and 18 Ω deionized water. For the blank diluent, 100 μL of 18 Ω deionized water was used to replace the sample serum. Sixty-two elements in the serum were analyzed using the iCAP Qc ICP-MS system (Agilent 7700x ICP-MS, USA) of Shanghai Biotree Biotech Co., Ltd. The response of the calibration curve to the measured concentration was evaluated for goodness of fit. The R values of the correlation coefficients of the regression equations are all greater than 0.99, except for Ge, which was 0.98 (Table S1). The limit of detection (LOD) was calculated based on the average of 10 consecutive measurements of the blank diluent. Concentrations of elements below the LOD were imputed as LOD/2. Elements with a detection rate of less than 50% (wolfram and samarium) were categorized as "undetectable" (Table S2). Coefficients of variation varied with limits around 10% and met the required standards. All concentrations showed a right-skewed distribution and were loge transformed prior to statistical analysis.

Statistical analysis

Normality of distribution was assessed using the Kolmogorov-Smirnov test. Continuous variables were expressed as mean ± standard deviation or median [interquartile range (IQR)]. Differences between age groups were determined using the ANOVA test or the KruskalWallis test for continuous variables, depending on the data distribution. The chi-squared test was used to compare categorical variables presented as frequencies. To identify potential binary covariates, the enrolled participants were divided into two groups according to sex, alcohol consumption, smoking, hypertension, and cerebral infarction. Student t-test was used to compare the difference in age between the two groups. Conditional permutation for variable importance via random forest analysis was used to identify important elements most strongly associated with chronological age. Variables were ranked in descending order based on the importance score with higher scores indicating greater importance. A sliding window of nested random forest was used to calculate out-of-bag (OOB) errors. The element model with the minimum OOB error corresponded to a certain number of elements, which were selected for further analysis. Multivariate linear regression analysis was then performed to explore the potential linear relationship between the concentration of each element and age. Beta and 95% confidence intervals (CIs) were calculated. To further assess the potential nonlinear relationship of individual elements with age, restricted cubic spline was performed adjusting for covariates with 3 knots at the 25th, 50th, and 75th percentiles, with the value at the 50th percentile used as the reference. False discovery rate (FDR) was used to control for multiple comparisons, and P after FDR ≤ 0.05 was considered statistically significant. Pairwise interaction analyses of linear regression were performed to examine the correlations among 5 elements. Pearson correlation analysis was used to evaluate the relationships between selected elemental concentrations and laboratory indices. We also constructed an elemental risk score (ERS) to develop a potential approach for assessing elemental age. The ERS was calculated as the sum of the standardized concentrations of the selected elements multiplied by their respective coefficients. The formula was ERS = ∑KK=1 βkEk, where βk denotes the respective coefficient, Ek denotes the element standardized concentrations, and K denotes the number of elements. Statistical analyses were performed using R software, version 3.5.1 (The R Foundation for Statistical Computing) and SPSS 22.0 (IBM, Chicago, USA).

Results

Study participant characteristics

The demographic characteristics of the study population are shown in Table 1. Of the 70 participants, 35 (50.0%) were men and 35 (50.0%) were women. The mean age was 65.5 years and the median age was 65.2 years. There were no significant differences in sex, body mass index (BMI), smoking, and alcohol consumption among the different age groups. In contrast, the prevalence of hypertension and cerebral infarction was higher in subjects older than 70 years. Regarding biochemical parameters, the levels of ALB, A/G, and TC were lower in subjects older than 70 years. In addition, there were significant decreases in red blood cell (RBC), hematocrit (HCT), hemoglobin (Hb), and lymphocyte percentage (LYMPH) in subjects older than 80 years.

Table 1
Characteristics of the enrolled participants.

Variables 31–40 years old (n = 10) 41–50 years old (n = 10) 51–60 years old (n = 10) 61–70 years old (n = 10) 71–80 years old (n = 10) 81–90 years old (n = 10) 91–100 years old (n = 10) P
Age, years 38.0 (32.8–38.3) 47.0 (45.8–47.3) 54.5 (51.0–57.3) 64.5 (63.0–66.5) 74.0 (72.0–76.3) 83.5 (81.8–84.0) 92.5 (91.0–94.3) 0.000
Male, n (%) 5 (50.0) 5 (50.0) 5 (50.0) 5 (50.0) 5 (50.0) 5 (50.0) 5 (50.0) 1.000
BMI, kg/m2 24.2 ± 2.9 22.9 ± 1.4 23.1 ± 2.5 22.9 ± 4.0 25.3 ± 2.5 22.1 ± 1.6 23.6 ± 4.3 0.275
Smokers, n (%) 2 (20.0) 2 (20.0) 2 (20.0) 2 (20.0) 0 (0) 1 (10.0) 0 (0) 0.564
Drinkers, n (%) 3 (30.0) 1 (10.0) 2 (20.0) 0 (0) 0 (0) 1 (10.0) 1 (10.0) 0.342
Hypertension, n (%) 1 (10.0) 2 (20.0) 4 (40.0) 3 (30.0) 6 (60.0) 6 (60.0) 9 (90.0) 0.005
Cerebral infarction, n (%) 1 (10.0) 2 (20.0) 2 (20.0) 4 (40.0) 7 (70.0) 7 (70.0) 6 (60.0) 0.014
FBG, mmol/L 5.02 ± 0.34 4.88 ± 0.59 5.35 ± 0.75 5.19 ± 0.67 5.27 ± 0.95 5.14 ± 0.95 4.73 ± 0.34 0.416
ALT, U/L 18.5 (12.5–25.5) 15.1 (13.4–21.6) 13.2 (11.2–24.7) 15.8 (12.7–22.6) 13.4 (11.6–16.7) 15.7 (10.0–22.7) 13.1 (9.0–20.5) 0.741
AST, UL 19.8 (16.0–30.1) 18.3 (15.8–22.5) 18.1 (15.0–20.9) 19.8 (18.0–23.2) 18.7 (15.8–21.4) 20.8 (17.3–26.2) 23.8 (18.0–28.6) 0.311
TBil, μmolL 7.7 ± 2.8 13.1 ± 3.6 11.6 ± 5.5 11.0 ± 5.0 10.9 ± 5.2 9.5 ± 6.1 10.8 ± 3.2 0.255
SCr, μmol/L 59.6 ± 13.1 63.9 ± 12.4 63.9 ± 12.3 60.5 ± 9.6 70.0 ± 15.7 78.2 ± 22.4 70.5 ± 17.6 0.099
BUN, mmol/L 4.49 ± 1.28 5.04 ± 1.25 5.04 ± 1.31 4.93 ± 0.61 5.94 ± 1.01 6.01 ± 1.77 5.07 ± 1.88 0.148
TP, g/L 77.5 (71.4–78.7) 69.1 (65.9–76.3) 72.9 (64.4–80.0) 68.9 (63.8–78.6) 71.5 (65.0–79.0) 64.4 (62.5–70.1) 67.1 (64.8–70.8) 0.106
ALB, g/L 49.2 (47.9–50.8) 47.1 (42.4–48.5) 45.0 (40.8–50.9) 44.7 (41.0–50.2) 45.3 (42.9–46.4) 40.2 (37.2–41.9) 38.8 (34.8–41.5) 0.000
GLOB, g/L 28.6 (23.9–29.6) 22.7 (20.4–25.4) 29.6 (27.8–40.8) 25.2 (21.2–28.2) 26.5 (23.4–29.1) 26.5 (23.7–29.2) 29.8 (24.3–32.7) 0.008
A/G 1.70 (1.68–1.88) 1.91 (1.78–2.23) 1.80 (1.50–2.05) 1.85 (1.60–2.11) 1.70 (1.58–1.85) 1.55 (1.42–1.71) 1.25 (1.18–1.53) 0.000
TC, mmol/L 4.38 (4.03–5.22) 4.55 (4.24–4.96) 5.25 (4.93–5.66) 5.01 (3.97–6.42) 4.52 (4.08–5.18) 4.36 (4.11–4.66) 3.91 (3.22–4.44) 0.018
TG, mmol/L 1.22 (0.83–3.24) 1.08 (0.70–1.53) 1.11 (0.97–2.15) 1.43 (1.08–1.96) 1.01 (0.86–1.68) 0.82 (0.73–1.28) 0.99 (0.77–1.22) 0.159
LDL-C, mmol/L 2.75 ± 0.71 2.91 ± 0.57 2.88 ± 0.68 2.81 ± 0.67 2.75 ± 0.57 2.44 ± 0.61 2.24 ± 0.60 0.183
HDL-C, mmol/L 1.51 ± 0.54 1.54 ± 0.49 1.87 ± 0.77 1.61 ± 0.62 1.53 ± 0.16 1.49 ± 0.21 1.37 ± 0.39 0.454
UA, μmol/L 316.2 (251.5–363.2) 283.0 (209.3–361.4) 337.8 (272.7–360.4) 247.3 (228.9–301.1) 299.5 (233.9–390.3) 273.2 (244.8–334.6) 306.6 (246.7–349.2) 0.628
WBC, 109 cells/L 5.45 (5.16–6.76) 5.31 (4.35–6.11) 5.35 (4.55–5.80) 5.14 (4.40–7.12) 5.00 (4.60–6.13) 5.86 (4.02–7.07) 5.45 (4.23–7.19) 0.911
RBC, 109 cells/L 4.60 (4.03–4.95) 4.57 (4.21–4.68) 4.41 (4.13–5.05) 4.72 (4.55–4.84) 4.78 (4.21–4.95) 3.94 (3.52–4.31) 3.82 (3.67–4.05) 0.001
HCT, L/L 40.9 ± 3.6 41.2 ± 1.9 41.2 ± 5.0 41.9 ± 3.0 41.6 ± 3.4 37.0 ± 4.7 35.9 ± 2.4 0.001
Hb, g/L 137.5 (127.0–150.3) 135.0 (130.8–144.5) 142.5 (131.5–148.3) 138.5 (127.3–141.5) 141.5 (124.5–148.5) 119.0 (109.5–132.5) 121.0 (117.8–127.3) 0.005
PLT, 109 cells/L 239.7 ± 49.1 187.3 ± 44.9 216.0 ± 75.5 224.1 ± 43.0 174.9 ± 55.8 189.7 ± 50.4 194.1 ± 50.2 0.097
MONO, 109 cells/L 0.34 ± 0.07 0.33 ± 0.07 0.36 ± 0.11 0.40 ± 0.08 0.35 ± 0.07 0.44 ± 0.11 0.61 ± 0.23 0.000
MONO, % 5.85 (5.33–6.90) 6.80 (5.45–7.25) 6.11 (5.48–7.51) 7.20 (6.02–8.51) 6.85 (6.28–7.41) 7.30 (6.78–9.73) 9.90 (9.28–11.10) 0.000
LYMPH, 109 cells/L 1.83 ± 0.34 1.84 ± 0.42 1.94 ± 0.82 1.72 ± 0.55 1.49 ± 0.41 1.42 ± 0.66 1.45 ± 0.61 0.209
LYMPH, % 32.4 ± 7.3 35.5 ± 5.1 34.1 ± 8.0 31. 3± 9.6 29.2 ± 6.5 25.4 ± 7.6 25.5 ± 8.1 0.018

Note: A/G = albumin-globulin ratio; ALB = albumin; ALT = alanine transaminase; AST = aspartate aminotransferase; ASMI = appendicular skeletal muscle mass index; BMI = body mass index; BUN = blood urea nitrogen; CA-199 = carbohydrate antigen 199; FBG = fasting blood glucose; GLOB = globulin; Hb = hemoglobin; HCT = hematocrit; HDL-C = high-density lipoprotein cholesterol; LDL-C = low-density lipoprotein cholesterol; LYMPH = lymphocyte; MONO = monocyte; PLT = platelet; RBC = red blood cell; SCr = serum creatinine; TBil = total bilirubin; TC = total cholesterol; TG = triglyceride; TP = total protein; UA = uric acid; WBC = white blood cell.


Elementomics profiling

We quantified 62 elements by ICP-MS, including 5 alkali metals, 5 alkaline earth metals, 5 metalloids, 24 transition metals, 6 post-transition metals, 14 lanthanoids, 2 actinoids, and 1 other nonmetal (Figure 1A). The distributions of elemental exposure levels and the heatmap of different age groups are shown in Table S2 and Figure S1, respectively. OOB error analysis showed the top 13 elements that might be associated with age (Figure 1B-C). Further linear regression analysis and restricted cubic spline analysis identified 5 age-associated elements, including lithium (Li) [β (95 %CI) = 0.448 (14.02, 36.32), P < 0.001, Plinear FDR < 0.001], boron (B) [β (95 %CI) = 0.289 (3.04, 20.02), P = 0.009, Plinear FDR = 0.022], calcium (Ca) [β (95 %CI) = -0.271 (-189.72, -30.69), P = 0.007, Plinear FDR = 0.022], titanium (Ti) [β (95 %CI) = -0.332 (-49.44, -12.11), P = 0.002, Plinear FDR = 0.010] and selenium (Se) [β (95 %CI) = -0.296 (-38.12, -8.06), P = 0.003, Pnonlinear FDR = 0.008] (Table S3). With increasing age, the levels of Li and B increased, whereas the levels of Ca and Ti decreased (Figure 1D and Figure 2A-D). Interestingly, the concentration of Se was elevated before 60 years of age, but decreased thereafter (Figure 1D and Figure 2E). Concentrations of 5 identified elements in different age groups are presented (Table S4). The interaction analysis was performed to explore the possible interactive effects of Li, B, Ca, Ti, Se on the association with age. However, no significant correlation was observed among the above five elements (Table S5).

Figure 1. Periodic table of elements and elements identified by random forest. (A) Periodic table of elements, with detected elements marked in red. (B) Elements identified by random forest. The bar chart illustrates the variable importance measures of 62 elements. (C) The scatter plot illustrates the out-of-bag errors of the detected elements. The minimum OOB error corresponds to the first 13 elements. (D) Heatmap of 5 selected elements in different age groups.

Figure 2. Relationship between age and concentrations of 5 identified elements. Multiple linear regression graphs and restricted cubic spline curves were used to analyze the correlation between age and concentration of Li (A), B (B), Ca (C), Ti (D), and Se (E).

Correlation between candidate elements and laboratory measurements

Pearson correlation analysis showed that the concentration of serum Ca was positively correlated with the levels of total protein (r = 0.584, P = 0.000), ALB (r = 0.551, P = 0.001). The concentration of serum Se was positively correlated with the level of HCT (r = 0.472, P = 0.020) (Table S6).

Construction of ERS

After fitting a multivariate linear regression between age and 5 candidate elements, the age prediction formula was established as the ERS. The ERS was calculated as the sum of the standardized concentrations of the selected elements multiplied by their respective coefficients (Table S7) as follows: ERS=4.5522 × Li + 6.0575 × B - 4.9990 × Ca - 7.0403 × Ti - 0.8849 × Se, which means that higher ERS represents older age.

Discussion

The identification of reliable biomarkers of aging is one of the major research goals in geroscience. Although numerous studies have investigated the role of age-related biomarkers in degenerative diseases [23], the relationship between elementomics and senescence has received little attention. To address this gap, our study measured comprehensive serum elemental exposure profiles to investigate the association of elemental changes with age.
Aging is an inevitable process that affects all organisms over time and occurs at multiple levels, including the molecular, cellular, organ, and organismal levels [24]. It has long been accepted that aging can be attributed to the accumulation of reactive oxygen species (ROS), DNA damage, mitochondrial dysfunction, impaired antioxidant defense, damaged autophagy process, loss of proteostasis, and telomere shortening, etc. [25]. During the aging process, trace and macroelements, which are critical regulators of metabolic and physiological pathways, are altered and can influence oxidative and inflammatory processes [23, 26]. For example, some trace elements (such as Se, Zn, Mn) have the ability to reduce oxidative damage or enhance repair capacity by acting as essential cofactors for antioxidant enzymes and the different types of glutathione peroxidases, and therefore may play a crucial role in the aging process [27, 28]. Our study identified 5 elements as the most important predictors of chronological age. Our analyses showed that 4 elements had a linear relationship with age. Serum concentrations of Li and B appeared to be positively associated with increasing age, while Ca and Ti showed a decrease with age. Se levels were elevated before 60 years of age and showed a decline thereafter. The highlight of this study is that we constructed an integrated predictive model including Li, B, Ca, Ti, and Se to generate ERS, which could potentially serve as a tool for predicting senility.
Essential trace elements and major elements play an indispensable role in maintaining human health by participating in various metabolic processes and signaling pathways [29]. Among these elements, Se is a trace element essential for several metabolic processes, including thyroid hormone metabolism, antioxidant defense systems, and immune function [30]. Se is absorbed in the form of oranic (selenomethionine and selenocysteine) or inorganic (selenate and selenite) by erythrocytes from the gastrointestinal tract and subsequently transported by plasma to tissues and cells [31]. A previous study showed that serum selenium levels were positively correlated with hematocrit (P <0.001, r = 0.215) in 1000 older adults [32]. Consistently, we found that Se concentration was positively correlated with the level of HCT, suggesting that the absorption of Se decreases with aging. Our results showed that Se concentration was increased before 60 years of age, but decreased thereafter. One study reported that centenarians (91-110 years) had lower Se levels compared with elderly subjects (60-90 years) [33]. However, another study showed that centenarians (≥ 100 years) had much higher Se levels compared with elderly controls [34]. The discrepancy may be attributed to the different comorbidity status between different study populations, as decreased Se levels have been associated with cognitive decline and hyperglycemia in the elderly [35, 36]. Interestingly, a previous study found that Se concentration was increased in the thyroids of old men, accompanied by a decrease in B and Li, indicating a redistribution of trace elements with aging [37]. Therefore, these results may indicate the importance of Se supplementation at an appropriate age with an appropriate dose. Further studies are needed to explore the exact effects and the underlying mechanism of Se on the aging process.
Similar to Se, B is absorbed from the gastrointestinal tract and excreted in the urine [38]. Boron has been shown to be beneficial for bone growth and maintenance, central nervous system function, and regulation of inflammatory response [39]. Our study found a positive correlation between aging and serum B concentration. In men, serum B increased and reached a plateau between 50 and 69 years of age, followed by a gradual increase after 70 years of age. In women, a gradual increase in serum B concentration was observed up to the age of 70 years. Our results were consistent with previous studies showing a positive correlation between age and blood B levels [40, 41]. Although studies have shown some benefits of higher B status in the immune system, embryonic development, brain function, liver development, osteoporosis, cancer therapy, and wound healing, high-dose boron showed opposite effects [38]. Therefore, additional data are needed to elucidate the appropriate supplemental amount of B for human health as well as its mechanism of action.
Li has been used for decades to treat mood disorders, including bipolar disorder, anxiety, and depression [42]. A previous study in a Japanese population found that the concentration of Li in drinking water was associated with lower mortality [43]. Here, we showed that Li levels decreased with increasing age. Consistent with this, another study reported that younger subjects (mean age = 13.4 years) had a lower brain-to-serum Li concentration ratio than adults (mean age = 37.3 years) with bipolar disorder, indicating a decrease in serum Li in the adults [44]. Recent preclinical studies have shown that lithium can protect against various forms of oxidative and xenobiotic stress, thereby extending the lifespan of fission yeast, C. elegans, and drosophila [45-49]. However, the precise effects of Li as a biomarker and intervention for human aging need to be further confirmed.
As a heavy metal, Ti can enter the bloodstream from the respiratory and gastrointestinal tracts, as well as from titanium-containing implants [50]. Once in the blood, Ti quickly dissociates and transforms into titanium dioxide, which is then transported to various tissues throughout the body by a protein called serum transferrin [51]. Within cells, Ti can increase the production of ROS, leading to oxidative stress, apoptosis, and the induction of inflammation [52]. Accumulation of Ti in the nervous system may contribute to pathological damage and the development of degenerative diseases, including Alzheimer's disease and Parkinson's disease [53, 54]. In aged rats, titanium dioxide nanoparticles promoted the disruption of the blood-brain barrier, resulting in neuronal dysfunction and cognitive impairment, suggesting a detrimental effect of increased Ti on the development of age-related diseases [55]. However, here we found that Ti levels decreased with increasing age. In the body, Ti is rapidly eliminated from the blood and accumulates in tissues with a highly perfused reticuloendothelial system, such as the liver and spleen [56-58]. Therefore, we hypothesized that decreased circulating Ti concentration may be attributed to increased ectopic accumulation of Ti in aging organs such as the brain. Future studies are needed to investigate the distribution of Ti in the body and its effects on the function of different organs during the aging process.
Ca plays a vital role in skeletal and muscle maintenance, hormone secretion, nerve impulse transmission, and vascular integrity and activity [59]. Ca is mainly absorbed in the small intestine, and calcium homeostasis is maintained by parathyroid hormone and calcitonin [60]. Ca2+ serves as a multifunctional second messenger that fine-tunes a variety of physiological and pathological processes within the cell [61]. Changes in Ca levels and the Ca2+ signaling pathway have also been associated with accelerated aging [61]. A variety of senescence stimuli (oxidative stress, telomere attrition, genotoxic agents, etc.) can stimulate Ca2+ influx through cell membrane calcium channels [62-64]; however, along with Ca2+ overload, inefficient coupling of mitochondrial respiration can result in the production of excess reactive oxygen species, leading to mitochondrial damage, DNA damage, and ultimately apoptosis [63]. Several studies have suggested that serum 1,25(OH)2D levels decrease with age, accompanied by impaired Ca absorption, leading to a defect in intestinal Ca absorption [65-67], which may explain our finding of decreased serum Ca levels with age. In addition, our finding of a positive correlation between calcium concentration and total protein as well as serum albumin is consistent with previous studies [68]. Calcium supplementation has been recommended for the prevention of age-related diseases such as osteoporosis [69]. However, the adverse effects of calcium supplementation have also been reported, especially the formation of kidney stones [69]. Therefore, further studies are needed to explore the appropriate dosage of Ca that may have the ability of anti-aging.
There are several potential limitations to this study. First, the number of subjects was insufficient and limited to the area of Jiangsu Province, which may limit the universality of the study results. To improve the quality of research, it is recommended to increase the sample size and recruit people from different locations for further study. Second, our study did not take into account the dietary structure, eating habits, nutritional status, and drinking water source of the participants, which may lead to relatively inaccurate results. Therefore, a more comprehensive further study is needed to confirm our findings. Third, it was difficult to exclude the possibility of causality bias due to the crosssectional nature of this study. In addition, due to the complex biological and molecular mechanisms involved in aging, it may be difficult to identify a single biomarker as a valid measure of healthy aging. Therefore, it is of utmost importance to conduct prospective studies to validate the effect of our ERS model in reflecting aging. Finally, our findings were not externally validated. The ERS needs to be validated by future well-designed experimental studies before it can be used as an indicator of aging in other studies.

Conclusions

This study identified five age-related elements in serum and constructed an ERS that may reflect the chronological age. Our findings emphasize the importance of assessing the changes in elements as well as their effects on the aging process.

Declarations

Funding

This work was supported by grants from the National Key Research and Development Plan of China (No. 2020YFC2008505 to Xiang Lu), the National Natural Science Foundation of China (No. 81970217 to Wei Gao and 81970218 to Xiang Lu), and the Jiangsu Commission of Health (No. LR2022004 and No. LKZ2023005 to Wei Gao).

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Author Contributions

Wei Gao and Xiang Lu conceived and designed the study. Kai-Yue Jia and Yan-Ru Li carried out the experiments. Hui-Xian Sun and Can Zhao analyzed and interpreted the data. Kai-Yue Jia and HuiXian Sun drafted the manuscript. Wei Gao, Xiang Lu, and Yan-Ru Li revised the manuscript. All authors read and approved the final version of the manuscript.

Data availability

The data will be available from the corresponding author upon reasonable request.

Ethics approval

The study was conducted in accordance with the tenets of the Declaration of Helsinki and was approved by the Ethics Committee of Sir Run Run Hospital, Nanjing Medical University (approval number 2019-SRS041).

Consent to participate

Informed consent was obtained from all individual participants included in the study.

References

1. Pal S, & Tyler JK. Epigenetics and aging. Sci Adv, 2016, 2(7): e1600584.[Crossref]

2. Fang EF, Scheibye-Knudsen M, Jahn HJ, Li J, Ling L, Guo H, et al. A research agenda for aging in China in the 21st century. Ageing Research Reviews, 2015, 24: 197-205. [Crossref]

3. Sun R, Cao H, Zhu X, Liu J-P, & Dong E. Current aging research in China. Protein & Cell, 2015, 6(5): 314-321. [Crossref]

4. Seals DR, Justice JN, & LaRocca TJ. Physiological geroscience: targeting function to increase healthspan and achieve optimal longevity. The Journal of Physiology, 2016, 594(8): 2001-2024. [Crossref]

5. Liang YD, Liu Q, Du MH, Liu Z, Yao SM, Zheng PP, et al. Urinary 8-oxo-7,8-dihydroguanosine as a potential biomarker of frailty for elderly patients with cardiovascular disease. Free Radic Biol Med, 2020, 152: 248-254. [Crossref]

6. Ahadi S, Zhou W, Schussler-Fiorenza Rose SM, Sailani MR, Contrepois K, Avina M, et al. Personal aging markers and ageotypes revealed by deep longitudinal profiling. Nat Med, 2020, 26(1): 83-90. [Crossref]

7. Mounicou S, Szpunar J, & Lobinski R. Metallomics: the concept and methodology. Chem Soc Rev, 2009, 38(4): 1119-1138. [Crossref]

8. Rono JK, Sun D, & Yang ZM. Metallochaperones: a critical regulator of metal homeostasis and beyond. Gene, 2022, 822: 146352. [Crossref]

9. Haraguchi H. Metallomics: the history over the last decade and a future outlook. Metallomics, 2017, 9(8): 1001-1013. [Crossref]

10. Jomova K, Makova M, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, et al. Essential metals in health and disease. Chem Biol Interact, 2022, 367: 110173. [Crossref]

11. Squadrone S, Brizio P, Abete MC, & Brusco A. Trace elements profile in the blood of Huntington' disease patients. J Trace Elem Med Biol, 2020, 57: 18-20. [Crossref]

12. Yu HN, Shen SR, & Yin JJ. Effects of metal ions, catechins, and their interactions on prostate cancer. Crit Rev Food Sci Nutr, 2007, 47(8): 711-719. [Crossref]

13. Costello LC, & Franklin RB. A comprehensive review of the role of zinc in normal prostate function and metabolism; and its implications in prostate cancer. Arch Biochem Biophys, 2016, 611: 100-112. [Crossref]

14. Zimta AA, Schitcu V, Gurzau E, Stavaru C, Manda G, Szedlacsek S, et al. Biological and molecular modifications induced by cadmium and arsenic during breast and prostate cancer development. Environ Res, 2019, 178: 108700. [Crossref]

15. Jankowska EA, von Haehling S, Anker SD, Macdougall IC, & Ponikowski P. Iron deficiency and heart failure: diagnostic dilemmas and therapeutic perspectives. Eur Heart J, 2013, 34(11): 816-829. [Crossref]

16. Rakic A, Milovanovich ID, Trbovich AM, Stefanović S, Nikolić D, Janković S, et al. Trace elements in different tissues in aging rats. J Trace Elem Med Biol, 2020, 62: 126604. [Crossref]

17. Aydemir D, Salman N, Karimzadehkhouei M, Alaca BE, Turan B, & Ulusu NN. Evaluation of the effects of aging on the aorta stiffness in relation with mineral and trace element levels: an optimized method via custom-built stretcher device. Biological Trace Element Research, 2021, 199(7): 2644-2652. [Crossref]

18. Shimamura T, Iijima S, Hirayama M, Iwashita M, Akiyama S, Takaku Y, et al. Age-related effects of major and trace element concentrations in rat liver and their mutual relationships. J Trace Elem Med Biol, 2013, 27(4): 286-294. [Crossref]

19. Zeng HL, Li H, Lu J, Guan Q, & Cheng L. Assessment of 12 metals and metalloids in blood of general populations living in Wuhan of China by ICP-MS. Biol Trace Elem Res, 2019, 189(2): 344-353. [Crossref]

20. Kim HJ, Lim HS, Lee KR, Choi MH, Kang NM, Lee CH, et al. Determination of trace metal levels in the general population of Korea. Int J Environ Res Public Health, 2017, 14(7): 702-712. [Crossref]

21. Zhang P, Georgiou CA, & Brusic V. Elemental metabolomics. Brief Bioinform, 2018, 19(3): 524-536. [Crossref]

22. Laur N, Kinscherf R, Pomytkin K, Kaiser L, Knes O, & Deigner HP. ICP-MS trace element analysis in serum and whole blood. PLoS One, 2020, 15(5): e0233357. [Crossref]

23. Wagner KH, Cameron-Smith D, Wessner B, & Franzke B. Biomarkers of aging: from function to molecular biology. Nutrients, 2016, 8(6): 338-349. [Crossref]

24. Moskalev A, Guvatova Z, Lopes IA, Beckett CW, Kennedy BK, De Magalhaes JP, et al. Targeting aging mechanisms: pharmacological perspectives. Trends Endocrinol Metab, 2022, 33(4): 266-280. [Crossref]

25. da Silva PFL, & Schumacher B. Principles of the molecular and cellular mechanisms of aging. J Invest Dermatol, 2021, 141(4s): 951-960. [Crossref]

26. Khan SS, Singer BD, & Vaughan DE. Molecular and physiological manifestations and measurement of aging in humans. Aging Cell, 2017, 16(4): 624-633. [Crossref]

27. Méplan C. Trace elements and ageing, a genomic perspective using selenium as an example. J Trace Elem Med Biol, 2011, 25 Suppl 1: S11-16. [Crossref]

28. Lossow K, Kopp JF, Schwarz M, Finke H, Winkelbeiner N, Renko K, et al. Aging affects sex- and organ-specific trace element profiles in mice. Aging (Albany NY), 2020, 12(13): 13762-13790. [Crossref]

29. Baudry J, Kopp JF, Boeing H, Kipp AP, Schwerdtle T, & Schulze MB. Changes of trace element status during aging: results of the EPIC-Potsdam cohort study. European Journal of Nutrition, 2019, 59(7): 3045-3058. [Crossref]

30. Mojadadi A, Au A, Salah W, Witting P, & Ahmad G. Role for selenium in metabolic homeostasis and human reproduction. Nutrients, 2021, 13(9): 3256-3265. [Crossref]

31. Mehdi Y, Hornick JL, Istasse L, & Dufrasne I. Selenium in the environment, metabolism and involvement in body functions. Molecules, 2013, 18(3): 3292-3311. [Crossref]

32. He Y, Yu J, Yu N, Chen R, Wang S, Wang Q, et al. Association between the ratios of selenium to several elements and mild cognitive impairment in the elderly. Biol Trace Elem Res, 2023, 201(9): 4286-4297. [Crossref]

33. Savarino L, Granchi D, Ciapetti G, Cenni E, Ravaglia G, Forti P, et al. Serum concentrations of zinc and selenium in elderly people: results in healthy nonagenarians/centenarians. Exp Gerontol, 2001, 36(2): 327-339. [Crossref]

34. Alis R, Santos-Lozano A, Sanchis-Gomar F, Pareja-Galeano H, Fiuza-Luces C, Garatachea N,et al. Trace elements levels in centenarian ‘dodgers'. J Trace Elem Med Biol, 2016, 35: 103-106. [Crossref]

35. Akbaraly TN, Arnaud J, Rayman MP, Hininger-Favier I, Roussel AM, Berr C, et al. Plasma selenium and risk of dysglycemia in an elderly French population: results from the prospective epidemiology of vascular ageing study. Nutr Metab (Lond), 2010, 7: 21-33. [Crossref]

36. Akbaraly TN, Hininger-Favier I, Carrière I, Arnaud J, Gourlet V, Roussel AM, et al. Plasma selenium over time and cognitive decline in the elderly. Epidemiology, 2007, 18(1): 52-58. [Crossref]

37. Zaichick V, & Zaichick S. Associations between age and 50 trace element contents and relationships in intact thyroid of males. Aging Clin Exp Res, 2018, 30(9): 1059- 1070. [Crossref]

38. Khaliq H, Juming Z, & Ke-Mei P. The physiological role of boron on health. Biol Trace Elem Res, 2018, 186(1): 31- 51. [Crossref]

39. Nielsen FH. Micronutrients in parenteral nutrition: boron, silicon, and fluoride. Gastroenterology, 2009, 137(5 Suppl): S55-60. [Crossref]

40. Usuda K, Kono K, & Yoshida Y. Serum boron concentration from inhabitants of an urban area in Japan. Reference value and interval for the health screening of boron exposure. Biol Trace Elem Res, 1997, 56(2): 167-178. [Crossref]

41. Hasbahceci M, Cipe G, Kadioglu H, Aysan E, & Muslumanoglu M. Reverse relationship between blood boron level and body mass index in humans: does it matter for obesity? Biological Trace Element Research, 2013, 153(1): 141-144. [Crossref]

42. Fountoulakis KN, Tohen M, & Zarate CA, Jr. Lithium treatment of bipolar disorder in adults: a systematic review of randomized trials and meta-analyses. Eur Neuropsychopharmacol, 2022, 54: 100-115. [Crossref]

43. Zarse K, Terao T, Tian J, Iwata N, Ishii N, & Ristow M. Low-dose lithium uptake promotes longevity in humans and metazoans. Eur J Nutr, 2011, 50(5): 387-389. [Crossref]

44. Moore CM, Demopulos CM, Henry ME, Steingard RJ, Zamvil L, Katic A, et al. Brain-to-serum lithium ratio and age: an in vivo magnetic resonance spectroscopy study. Am J Psychiatry, 2002, 159(7): 1240-1242. [Crossref]

45. Sofola-Adesakin O, Castillo-Quan JI, Rallis C, Tain LS, Bjedov I, Rogers I, et al. Lithium suppresses Aβ pathology by inhibiting translation in an adult Drosophila model of Alzheimer's disease. Front Aging Neurosci, 2014, 6: 190- 200. [Crossref]

46. McColl G, Killilea DW, Hubbard AE, Vantipalli MC, Melov S, & Lithgow GJ. Pharmacogenetic analysis of lithiuminduced delayed aging in Caenorhabditis elegans. J Biol Chem, 2008, 283(1): 350-357. [Crossref]

47. Castillo-Quan JI, Li L, Kinghorn KJ, Ivanov DK, Tain LS, Slack C, et al. Lithium promotes longevity through GSK3/ NRF2-dependent hormesis. Cell Rep, 2016, 15(3): 638- 650. [Crossref]

48. Fries GR, Zamzow MJ, Colpo GD, Monroy-Jaramillo N, Quevedo J, Arnold JG, et al. The anti-aging effects of lithium in lymphoblastoid cell lines from patients with bipolar disorder and controls. Journal of Psychiatric Research, 2020, 128: 38-42. [Crossref]

49. Castillo-Quan Jorge I, Li L, Kinghorn Kerri J, Ivanov Dobril K, Tain Luke S, Slack C, et al. Lithium promotes longevity through GSK3/NRF2-dependent hormesis. Cell Reports, 2016, 15(3): 638-650. [Crossref]

50. Saxena M, Loza-Rosas SA, Gaur K, Sharma S, Pérez Otero SC, & Tinoco AD. Exploring titanium(IV) chemical proximity to iron(III) to elucidate a function for Ti(IV) in the human body. Coord Chem Rev, 2018, 363: 109-125. [Crossref]

51. Loza-Rosas SA, Saxena M, Delgado Y, Gaur K, Pandrala M, & Tinoco AD. A ubiquitous metal, difficult to track: towards an understanding of the regulation of titanium(iv) in humans. Metallomics, 2017, 9(4): 346-356. [Crossref]

52. Ma Y, Yu N, Lu H, Shi J, Zhang Y, Chen Z, et al. Titanium dioxide nanoparticles: revealing the mechanisms underlying hepatotoxicity and effects in the gut microbiota. Arch Toxicol, 2023, 97(8): 2051-2067. [Crossref]

53. Fu PP, Xia Q, Hwang H-M, Ray PC, & Yu H. Mechanisms of nanotoxicity: Generation of reactive oxygen species. Journal of Food and Drug Analysis, 2014, 22(1): 64-75. [Crossref]

54. Swiatkowska I, Martin N, & Hart AJ. Blood titanium level as a biomarker of orthopaedic implant wear. J Trace Elem Med Biol, 2019, 53: 120-128. [Crossref]

55. Disdier C, Chalansonnet M, Gagnaire F, Gaté L, Cosnier F, Devoy J, et al. Brain inflammation, blood brain barrier dysfunction and neuronal synaptophysin decrease after inhalation exposure to titanium dioxide nano-aerosol in aging rats. Sci Rep, 2017, 7(1): 12196. [Crossref]

56. Geraets L, Oomen AG, Krystek P, Jacobsen NR, Wallin H, Laurentie M, et al. Tissue distribution and elimination after oral and intravenous administration of different titanium dioxide nanoparticles in rats. Part Fibre Toxicol, 2014, 11: 30-42. [Crossref]

57. Fabian E, Landsiedel R, Ma-Hock L, Wiench K, Wohlleben W, & van Ravenzwaay B. Tissue distribution and toxicity of intravenously administered titanium dioxide nanoparticles in rats. Arch Toxicol, 2008, 82(3): 151-157. [Crossref]

58. Shinohara N, Danno N, Ichinose T, Sasaki T, Fukui H, Honda K, et al. Tissue distribution and clearance of intravenously administered titanium dioxide (TiO2) nanoparticles. Nanotoxicology, 2014, 8(2): 132-141. [Crossref]

59. Emkey RD, & Emkey GR. Calcium metabolism and correcting calcium deficiencies. Endocrinol Metab Clin North Am, 2012, 41(3): 527-556. [Crossref]

60. Li K, Wang XF, Li DY, Chen YC, Zhao LJ, Liu XG, et al. The good, the bad, and the ugly of calcium supplementation: a review of calcium intake on human health. Clin Interv Aging, 2018, 13: 2443-2452. [Crossref]

61. Madreiter-Sokolowski CT, Thomas C, & Ristow M. Interrelation between ROS and Ca(2+) in aging and age-related diseases. Redox Biol, 2020, 36: 101678. [Crossref]

62. Barbagallo M, Dominguez LJ, Licata G, & Resnick LM. Effects of aging on serum ionized and cytosolic free calcium: relation to hypertension and diabetes. Hypertension, 1999, 34(4 Pt 2): 902-906. [Crossref]

63. Ureshino RP, Rocha KK, Lopes GS, Bincoletto C, & Smaili SS. Calcium signaling alterations, oxidative stress, and autophagy in aging. Antioxid Redox Signal, 2014, 21(1): 123-137. [Crossref]

64. Ziegler DV, Vindrieux D, Goehrig D, Jaber S, Collin G, Griveau A, et al. Calcium channel ITPR2 and mitochondria–ER contacts promote cellular senescence and aging. Nature Communications, 2021, 12(1): 720-734. [Crossref]

65. Kinyamu HK, Gallagher JC, Balhorn KE, Petranick KM, & Rafferty KA. Serum vitamin D metabolites and calcium absorption in normal young and elderly free-living women and in women living in nursing homes. Am J Clin Nutr, 1997, 65(3): 790-797. [Crossref]

66. Kinyamu HK, Gallagher JC, Prahl JM, DeLuca HF, Petranick KM, & Lanspa SJ. Association between intestinal vitamin D receptor, calcium absorption, and serum 1,25 dihydroxyvitamin D in normal young and elderly women. J Bone Miner Res, 1997, 12(6): 922-928. [Crossref]

67. Nordin BE, Need AG, Morris HA, O'Loughlin PD, & Horowitz M. Effect of age on calcium absorption in postmenopausal women. Am J Clin Nutr, 2004, 80(4): 998- 1002. [Crossref]

68. Yang YY, Zhang D, Ma LY, Hou YF, Bi YF, Xu Y, et al. Association of famine exposure and the serum calcium level in healthy Chinese adults. Front Endocrinol (Lausanne), 2022, 13: 937380. [Crossref]

69. Cormick G, & Belizan JM. Calcium Intake and Health. Nutrients, 2019, 11(7): 1606-1612. [Crossref].

Supplementary

Table S1
Correlation coefficients in calibration curves.

Element Abbreviation R Element Abbreviation R Element Abbreviation R
Lithium 7Li 0.9999 Selenium 78Se 0.9993 Gadolinium 157Gd 0.9999
Beryllium 9Be 0.9994 Rubidium 85Rb 0.9998 Terbium 159Tb 0.9999
Boron 11B 0.9996 Strontium 88Sr 0.9995 Dysprosium 163Dy 0.9999
Sodium 23Na 0.9989 Yttrium 89Y 0.9988 Holmium 165Ho 0.9998
Magnesium 24Mg 0.9990 Zirconium 90Zr 0.9995 Erbium 166Er 0.9997
Aluminum 27Al 0.9991 Niobium 93Nb 0.9813 Thulium 169Tm 0.9998
Potassium 39K 0.9999 Molybdenum 95Mo 0.9994 Ytterbium 172Yb 0.9999
Calcium 44Ca 0.9994 Ruthenium 101Ru 1 Lutetium 175Lu 0.9998
Scandium 45Sc 0.9995 Silver 107Ag 0.9999 Hafnium 178Hf 1
Titanium 47Ti 0.9996 Cadmium 111Cd 0.9990 Tantalum 181Ta 0.9982
Vanadium 51V 0.9986 Tin 118Sn 0.9999 Wolfram 182W 0.9998
Chromium 52Cr 0.9998 Antimony 121Sb 0.9997 Osmium 189Os 1
Manganese 55Mn 0.9998 Tellurium 125Te 1 Iridium 193Ir 1
Iron 56Fe 0.9995 Cesium 133Cs 1 Platinum 195Pt 1
Cobalt 59Co 0.9996 Barium 137Ba 0.9998 Hydrargyrum 202Hg 0.9996
Nickel 60Ni 1 Lanthanum 139La 0.9999 Thallium 205Tl 0.9994
Copper 63Cu 1 Cerium 140Ce 0.9999 Lead 208Pb 0.9999
Zinc 66Zn 0.9996 Praseodymium 141Pr 0.9999 Bismuth 209Bi 0.9998
Gallium 71Ga 0.9999 Neodymium 146Nd 0.9999 Thorium 232Th 1
Germanium 72Ge 0.9784 Samarium 147Sm 0.9999 Uranium 238U 1
Arsenic 75As 0.9999 Europium 151Eu 0.9999

Table S2
Quality control and statistical summary of element exposure levels in serum samples.

Abbreviation Unit LOD N (%) of Conc. RSD (%) Median (Q1–Q3)
7Li μg/L 0.0004 0 1.25 2.255 (1.785-2.843)
9Be μg/L 0.0005 5 (7.14%) 1.17 0.020 (0.010-0.030)
11B μg/L 0.4200 0 0.73 24.720 (17.238-34.703)
23Na mg/L 0.3400 0 0.05 3260.000 (3167.000-3353.250)
24Mg μg/L 0.0220 0 0.09 20883.500 (20148.250-21661.750)
27Al μg/L 0.0280 0 1.71 116.210 (85.750-213.000)
39K mg/L 1.7000 0 0.70 310.320 (173.752-513.530)
44Ca mg/L 0.8100 0 0.05 93.331 (91.214-97.321)
45Sc μg/L 0.0010 0 1.42 1.115 (0.708-1.693)
47Ti μg/L 0.0140 0 0.25 82.314 (74.110-99.251)
51V μg/L 0.0015 0 0.27 4.795 (3.663-5.913)
52Cr μg/L 0.0110 0 1.42 2.385 (1.685-3.483)
55Mn μg/L 0.0031 0 0.62 14.350 (6.508-21.581)
56Fe μg/L 0.0810 0 0.39 1199.531 (827.243-1493.250)
59Co μg/L 0.0006 1 (1.42%) 0.55 0.171 (0.118-0.232)
60Ni μg/L 0.0030 0 0.60 3.195 (2.703-4.185)
65Cu μg/L 0.0055 0 0.18 997.521 (937.253-1120.511)
66Zn μg/L 0.1500 0 0.37 917.21 (794.510-1070.250)
71Ga μg/L 0.0005 2 (2.86%) 0.77 0.050 (0.038-0.091)
72Ge μg/L 0.0055 9 (12.85%) 0.87 0.033 (0.010-0.060)
75As μg/L 0.0025 8 (11.42%) 1.02 0.432 (0.128-0.815)
78Se μg/L 0.2500 0 0.24 41.322 (34.175-47.125)
85Rb μg/L 0.0010 0 0.60 285.511 (182.752-419.250)
86Sr μg/L 0.0008 0 0.24 42.623 (36.721-47.950)
89Y μg/L 0.0002 6 (8.57%) 0.97 19.422 (0.310-28.051)
90Zr μg/L 0.0004 0 0.81 8.951 (3.420-17.230)
93Nb μg/L 0.0017 0 0.73 0.620 (0.408-0.913)
95Mo μg/L 0.0150 0 0.39 2.121 (1.623-2.725)
101Ru μg/L 0.0013 7 (10.00%) 0.77 0.020 (0.010-0.030)
107Ag μg/L 0.0010 5 (7.14%) 2.06 0.035 (0.021-0.080)
111Cd μg/L 0.0010 1 (1.42%) 0.62 0.070 (0.051-0.103)
118Sn μg/L 0.0046 5 (7.14%) 1.45 0.371 (0.132-3.695)
121Sb μg/L 0.0007 0 0.41 4.923 (3.678-6.118)
125Te μg/L 0.0300 12 (17.14%) 0.89 0.515 (0.121-0.901)
133Cs μg/L 0.0001 0 0.37 0.992 (0.831-1.293)
137Ba μg/L 0.0019 0 0.71 5.661 (4.150-6.821)
139La μg/L 0.0002 0 0.40 0.175 (0.148-0.223)
140Ce μg/L 0.0003 0 0.39 0.312 (0.241-0.380)
141Pr μg/L 0.0001 2 (2.86%) 0.47 0.040 (0.030-0.051)
146Nd μg/L 0.0003 0 0.44 0.145 (0.108-0.18)
147Sm μg/L 0.0003 38 (64.29%) 1.53 0 (0-0.020)
151Eu μg/L 0.0001 10 (14.29%) 0.78 0.010 (0.010-0.020)
157Gd μg/L 0.0003 2 (2.86%) 0.56 0.080 (0.050-0.120)
159Tb μg/L 0.0001 8 (11.43%) 0.72 0.010 (0.010-0.020)
163Dy μg/L 0.0002 2 (2.86%) 0.67 0.065 (0.040-0.113)
165Ho μg/L 0.0001 7 (10.00%) 0.74 0.020 (0.011-0.030)
166Er μg/L 0.0004 5 (7.14%) 0.67 0.050 (0.030-0.071)
169Tm μg/L 0.0025 19 (27.14%) 0.93 0.010 (0.001-0.011)
172Yb μg/L 0.0002 1 (1.42%) 0.63 0.040 (0.031-0.070)
175Lu μg/L 0.0002 34 (48.57%) 1.28 0.011 (0-0.012)
178Hf μg/L 0.0001 0 0.86 2.580 (1.908-4.055)
181Ta μg/L 0.0008 0 0.52 0.181 (0.140-0.255)
182W μg/L 0.0030 48 (68.57%) 2.77 0.002 (0.002-0.343)
189Os μg/L 0.0004 0 2.08 0.025 (0.020-0.040)
193Ir μg/L 0.0001 0 0.93 0.021 (0.020-0.033)
195Pt μg/L 0.0005 1 (1.42%) 0.73 0.080 (0.031-0.130)
201Hg μg/L 0.0180 0 0.34 1.795 (1.580-2.201)
205Tl μg/L 0.0005 0 1.73 0.300 (0.201-0.690)
208Pb μg/L 0.0013 0 2.10 1.791 (1.365-2.325)
209Bi μg/L 0.0002 0 1.45 0.065 (0.040-0.158)
232Th μg/L 0.0003 0 1.73 0.435 (0.188-1.228)
238U μg/L 0.0003 6 (8.57%) 1.06 0.040 (0.010-0.061)

Note: Conc: concentration; LOD: limit of detection, calculated as 3 times the average of 10 consecutive measurements of the blank diluent; RSD: relative standard deviation to evaluate stability of the measurements.


Table S3
Results of multiple linear regression analysis and restricted cubic spline regression of the association of age with elements.

Element β 95%CI P Plinear FDR Pnonlinear Pnonlinear FDR
1Li 0.448 (14.02, 36.32) 0.000 0.000 0.183 0.362
3B 0.289 (3.04, 20.02) 0.009 0.022 0.118 0.362
4Na -0.168 (-161.39, 13.74) 0.097 0.140 0.153 0.362
7K 0.216 (0.67, 12.22) 0.029 0.064 0.264 0.381
8Ca -0.271 (-189.72, -30.69) 0.007 0.022 0.021 0.136
10Ti -0.332 (-49.44, -12.11) 0.002 0.010 0.507 0.599
14Fe -0.192 (-18.09, 0.82) 0.073 0.118 0.109 0.362
18Zn -0.200 (-30.69, 0.58) 0.059 0.109 0.195 0.362
22Se -0.296 (-38.12, -8.06) 0.003 0.013 0.001 0.008
23Rb 0.130 (-2.62, 11.86) 0.207 0.270 0.572 0.620
35Cs 0.085 (-6.94, 17.01) 0.404 0.437 0.227 0.368
43Gd -0.105 (-9.74, 3.1) 0.305 0.361 0.379 0.492
54Os -0.014 (-3.65, 3.2) 0.895 0.895 0.686 0.686

Note: Linear regression analysis and restricted cubic spline regression adjusted for three covariates including smoking, hypertension, and cerebral infarction.


Table S4
Concentrations of 5 identified elements in different age groups.

Variables 31–40 years old (n = 10) 41–50 years old (n = 10) 51–60 years old (n = 10) 61–70 years old (n = 10) 71–80 years old (n = 10) 81–90 years old (n = 10) 91–100 years old (n = 10) P
Li, μg/L 1.52 (1.28–1.89) 2.07 (1.39–3.00) 1.83 (1.65–1.98) 2.37 (2.14–2.59) 2.00 (2.25–2.92) 2.37 (1.77–3.62) 2.89 (2.42–3.39) 0.451
B, μg/L 18.1 (13.5–22.9) 14.3 (11.6–23.9) 23.6 (19.1–31.0) 26.5 (18.4–37.7) 37.3 (27.2–39.7) 29.5 (17.7–51.4) 31.8 (23.6–56.5) 0.047
Ca, mg/L 96.5 (93.0–98.3) 93.5 (93.1–97.3) 97.0 (92.8–97.5) 94.1 (90.0–98.2) 93.1 (91.8–98.3) 91.5 (88.8–97.3) 90.5 (86.5–92.3) 0.010
Ti, μg/L 95.1 (82.2–106.8) 82.5 (79.8–96.0) 82.5 (67.1–108.5) 93.5 (79.5–102.8) 82.1 (75.3–103.5) 73.3 (67.3–88.0) 76.0 (61.8–81.3) 0.051
Se, μg/L 43.2 ± 8.9 44.7 ± 6.7 42.7 ± 12.1 46.8 ± 10.7 46.0 ± 7.5 36.1 ± 4.9 30.9 ± 8.0 0.001

Table S5
Pairwise interaction of 5 identified elements.

Element Pinteraction
Li*B 0.2869
Li*Ca 0.9687
Li*Ti 0.7234
Li*Se 0.666
B*Ca 0.7232
B*Ti 0.0676
B*Se 0.6741
Ca*Ti 0.4234
Ca*Se 0.1067
Ti*Se 0.7531

Table S6
Correlation between 5 identified elements and laboratory measurements.

Element Laboratory measurement r P value P Bonferroni
Ca TP, g/L 0.584 0.000 0.000
Ca ALB, g/L 0.551 0.000 0.001
Se HCT, L/L 0.472 0.000 0.020
Se RBC, 109 cells/L 0.440 0.000 0.076
Ca HCT, L/L 0.441 0.000 0.081
Ca Hb, g/L 0.430 0.000 0.123
Ca RBC, 109 cells/L 0.415 0.000 0.211
Se MONO, % -0.411 0.000 0.221
Ti Hb, g/L 0.380 0.001 0.643
Ca MONO, % -0.374 0.002 0.840

Note: ALB = albumin; r = correlation coefficient; Hb = hemoglobin; HCT = hematocrit; LYMPH = lymphocyte; MONO = monocytes; RBC = white blood cell; TP = total protein.


Table S7
Correlation between 5 identified elements and laboratory measurements.

Element β 95%CI P
Li 4.552 (0.99, 8.12) 0.013
B 6.058 (2.43, 9.67) 0.001
Ca -4.999 (-8.29, -1.71) 0.004
Ti -7.040 (-10.43, -3.65) 0.001
Se -0.885 (-4.37, 2.60) 0.613

Figure S1. Heatmap of detected elements. Participants were divided into 7 groups as 10 years span of age.



Subscribe to receive issue release notifications
and newsletters from journals