Next Article in Journal
Development of a Digital Photographic Food Atlas as a Portion Size Estimation Aid in Japan
Previous Article in Journal
Bioactive Natural Compounds for Therapeutic and Nutraceutical Applications in Neurodegeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin Supplementation Protects against Nanomaterial-Induced Oxidative Stress and Inflammation Damages: A Meta-Analysis of In Vitro and In Vivo Studies

1
College of Textile and Clothing Engineering, Soochow University, 199 Ren-Ai Road, Suzhou 215123, China
2
Department of Health and Hazards Surveillance, Shinan District Center for Disease Control and Prevention, Qingdao 266072, China
3
Shanghai Jing Rui Yang Industrial Co., Ltd., 3188 Xiupu Road, Shanghai 200122, China
4
Shanghai Nutri-Woods Bio-Technology Co., Ltd., No. 333 Guiping Road, Shanghai 200233, China
5
Shanghai Pechoin Daily Chemical Co., Ltd., No. 518 Anyuan Road, Shanghai 200060, China
6
Shanghai Institute of Spacecraft Equipment, 251 Huaning Road, Shanghai 200240, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 26 April 2022 / Revised: 19 May 2022 / Accepted: 25 May 2022 / Published: 26 May 2022
(This article belongs to the Section Micronutrients and Human Health)

Abstract

:
The extensive applications of nanomaterials have increased their toxicities to human health. As a commonly recommended health care product, vitamins have been reported to exert protective roles against nanomaterial-induced oxidative stress and inflammatory responses. However, there have been some controversial conclusions in regards to this field of research. This meta-analysis aimed to comprehensively evaluate the roles and mechanisms of vitamins for cells and animals exposed to nanomaterials. Nineteen studies (seven in vitro, eleven in vivo and one in both) were enrolled by searching PubMed, EMBASE, and Cochrane Library databases. STATA 15.0 software analysis showed vitamin E treatment could significantly decrease the levels of oxidants [reactive oxygen species (ROS), total oxidant status (TOS), malondialdehyde (MDA)], increase anti-oxidant glutathione peroxidase (GPx), suppress inflammatory mediators (tumor necrosis factor-α, interleukin-6, C-reactive protein, IgE), improve cytotoxicity (manifested by an increase in cell viability and a decrease in pro-apoptotic caspase-3 activity), and genotoxicity (represented by a reduction in the tail length). These results were less changed after subgroup analyses. Pooled analysis of in vitro studies indicated vitamin C increased cell viability and decreased ROS levels, but its anti-oxidant potential was not observed in the meta-analysis of in vivo studies. Vitamin A could decrease MDA, TOS and increase GPx, but its effects on these indicators were weaker than vitamin E. Also, the combination of vitamin A with vitamin E did not provide greater anti-oxidant effects than vitamin E alone. In summary, we suggest vitamin E alone supplementation may be a cost-effective option to prevent nanomaterial-induced injuries.

1. Introduction

With the rapid development of nanotechnology in the last decade, nanomaterials have been omnipresent in industrial products, medicines, food, and cosmetics due to their unique chemical and physical properties [1,2,3]. These extensive applications make nanomaterials unavoidably enter human bodies through respiratory inhalation, dermal penetration, oral ingestion, or injection routes, which subsequently results in toxic damages on various organs and tissues (e.g., lung, liver, kidney, spleen, heart, testis, et al.) and contributes to the appearance of related diseases [4,5]. Therefore, it is urgently necessary to introduce some preventive strategies to antagonize the detrimental effects of nanomaterials on human health.
Although the toxicological mechanisms of nanomaterials have not been well elucidated, activations of oxidative stress and inflammation have been demonstrated to play important roles [6,7]. An et al. used a meta-analysis to observe that titanium dioxide (TiO2) nanoparticle (NP) exposure significantly increased the levels of oxidants (malonaldehyde, MDA; 8-hydroxy-2-deoxyguanosine; superoxide anion; hydrogen peroxide) and reduced the levels of anti-oxidants (superoxide dismutase, SOD; glutathione, GSH; glutathione peroxidase, GPx; catalase, CAT) in murine models compared with the controls [6]. In addition, to cause redox perturbation, Moradi et al. found that TiO2NP gavage promoted the expression of pro-inflammatory cytokine tumor necrosis factor (TNF-α) and nuclear factor kappaB (NF-κB) in the liver tissues of rats in comparison to a control group [8]. Elevated levels of MDA, TNF-α, interleukin-6 (IL-6), C-reactive protein (CRP), and decreased levels of GSH were also revealed in the rats that received zinc oxide (ZnO) [9,10] or gold NPs [11] compared to the controls. In vitro studies showed that carbon nanotube or NP treatment inhibited the viability and induced apoptotic cell death by increasing the levels of reactive oxygen species (ROS) [12,13,14]. These findings indicate that supplementation with compounds that have anti-oxidant and anti-inflammatory functions may represent a potential strategy to prevent nanomaterial-induced toxic damages.
Currently, vitamins are one of the most commonly recommended health care products in the medical market. Several studies have reported that vitamin intake can reduce oxidative stress and inhibit pro-inflammatory responses [15,16], indicating that there are potential protective effects of vitamin supplementation against tissue injuries induced by nanomaterials. This hypothesis had been confirmed by some authors: Bayat et al. detected that relative to the ZnONP-exposed group, administration with vitamin A, C, and E significantly reduced the total oxidant status (TOS), oxidative stress index (OSI) and increased the total anti-oxidant capacity (TAC) and activities of SOD, GPx, and CAT [10], which was accompanied by decreased liver damages pathologically [10] and DNA damages [17]. Moradi et al. reported that treatment with vitamin A and E caused significant reductions in MDA, TNF-α, and NF-κB, as well as liver function index (aspartate aminotransferase, AST) [8]. However, some controversial results were also reported. The study of Afshari-Kaveh et al. showed that treatment with vitamin A alone could not prevent TiO2 NP-induced decreases in TAC and OSI and increases in TOS [18]. Kong et al. demonstrated that there were no significant differences in SOD, GPx, and CAT between vitamin C-treated and NP-exposed groups [19]. AL-RASHEED et al. found that vitamin E could not significantly protect liver tissues from DNA damage (indicated by a decrease in tail length) compared with ZnONP-intoxicated rats [9]. Accordingly, the protective effects of vitamin supplementation against tissue injuries induced by nanomaterials remain inconclusive.
In this study, we aimed to conduct a meta-analysis based on in vivo and in vitro studies to comprehensively evaluate whether supplementation with vitamins would enhance cell viability, inhibit cell apoptosis, and prevent tissue injuries caused by nanomaterials through exerting anti-oxidant and anti-inflammatory activities.

2. Materials and Methods

2.1. Search Strategy

This meta-analysis was performed according to the preferred reporting items for systematic reviews and meta-analysis (PRISMA). Three electronic databases, including PubMed, EMBASE and Cochrane Library, were searched from inception to January 2022 to retrieve relevant studies. The search terms used were as follows: (“nanoparticle” OR “carbon nanotube” OR “graphene”) AND (“vitamin”) AND (“animal” OR “cells”) AND (toxicity). Additionally, the reference lists of the relevant studies and reviews were also manually checked to identify potential complements.

2.2. Inclusion and Exclusion Criteria

Studies were included based on the participants, interventions, comparisons, outcomes, and study design (PICOS) principles: (1) participants (P): murine or murine (human) cells; (2) intervention (I): the experimental group was treated with nanomaterials and vitamins; (3) comparison (C): the control group only received nanomaterial exposure; (4) outcomes (O): cell viability, apoptosis (caspase-3), DNA damage (tail length, tail DNA %), oxidative stress (ROS, MDA, TOS, TAC, OSI, SOD, GSH, GPx, CAT; Nrf2, nuclear factor E2-related factor 2), inflammation (TNF-α, NF-κB, IL-6, CRP, IgE), global health issues (body weight) and tissue damage (such as AST; ALT, alanine aminotransferase); and (5) study design (S): controlled trials.
Articles were excluded if they met the following criteria: (1) duplicate publications; (2) non-original research, such as abstracts, case reports, reviews, letter to the editor and comments; (3) data could not be extracted or combined with other studies; and (4) irrelevant to the study objectives. Two authors independently performed the literature selection and any disagreements were resolved by discussion with a third reviewer.

2.3. Data Extraction

The following data were independently extracted from each qualified article by two reviewers, including the first author, the publication year, country, nanomaterial type, vitamin type, cell or animal type, number per group, dosage of nanomaterials or vitamins, duration of treatment, sample source for analysis of outcomes and data of the experimental and control groups (mean ± standard deviation). A digitizing software (Engauge Digitizer (http://digitizer.sourceforge.net/, accessed on 23 March 2022) was used to extract the data presented in the graphs. Any discrepancies in the above information were resolved by discussion with a third reviewer.

2.4. Quality Assessment

The Toxrtool scale was used to evaluate the quality of in vitro studies [20,21], which consisted of 18 items. A score of 1 was assigned for each item if the article satisfied the criteria; 1; otherwise, the score of 0 was given for the articles. Studies with a Toxrtool score of ≥11 points were considered to be of high quality. The SYRCLE risk-of-bias tool was used to evaluate the quality of in vivo studies [22], which consisted of ten questions about selection, performance, detection, attrition, reporting and other bias in included articles. Items were divided into ‘low’, ‘high’ or ‘unclear’ risk of bias when they were labeled as yes, no, unknown to selected articles. The methodological quality was independently evaluated by two authors. Any divergence was resolved through discussion with a third reviewer.

2.5. Statistical Analysis

Meta-analyses were performed using STATA 15.0 software (STATA Corporation, College Station, TX, USA). The standardized mean difference (SMD) and 95% confidence interval (CI) were calculated to assess the intervention effects of vitamins. The heterogeneity between studies was examined by Cochrane’s Q-square test and I2 statistic. If significant heterogeneity was present (p < 0.1 and I2 > 50%), a random-effects model was used to analyze the combined effect size; otherwise, a fixed-effects model was employed. Subgroup analyses stratified by nanomaterial types, study durations, dosages of vitamins, sample sources and animal models were conducted for indicators with at least five data analyzed to determine whether these factors influenced the results. Publication bias was evaluated by Egger’s linear regression test. The trim-and-fill method was used to adjust the pooled SMDs if a significant publication bias was detected (p < 0.05). The robustness of the pooled conclusions was assessed using a sensitivity analysis based on stepwise removing one study at a time.

3. Results

3.1. Study Selection

A total of 1212 articles were initially retrieved through searching the electronic databases, of which 405 were duplicates. Titles and abstracts of 807 studies were reviewed and then 758 were excluded because they were irrelevant to the study’s objective. The full-texts of the remaining 49 studies were downloaded and read, after which 29 studies were eliminated for the following reasons: data were unavailable (n = 4); outcome indicators could not be combined with other studies (n = 6); the effects of some vitamin subtypes were reported only in one study (vitamin D [23] and vitamin K [24]); studies did not use mice or rats as animal models (n = 14); vitamins were mixed with other nutrients (n = 4). Finally, 19 studies were included in this meta-analysis (Figure 1) [8,9,10,11,12,13,14,17,18,19,25,26,27,28,29,30,31,32,33].

3.2. Study Characteristics

The basic characteristics of included articles are summarized in Table 1. Among 19 studies, seven performed in vitro experiments [12,13,14,30,31,32,33], 11 performed in vivo experiments [8,9,10,11,17,18,19,25,26,28,29] and one conducted both in vitro and in vivo experiments [27]. The in vitro studies were published between 2007 and 2019; four studies were conducted in China and the remaining four studies were, respectively, conducted in Iran, Japan, Saudi Arabia and the USA; there were four studies to, respectively, investigate the protective roles of vitamin E and vitamin C against the toxicities of various nanomaterials (including TiO2NPs; CoNPs, cobalt nanoparticles; AgNPs, silver nanoparticles; ZnONPs; NiONPs, nickel oxide nanoparticles; SWCNTs, single walled carbon nanotubes); the exposed cells included liver cells (human HepG2, mice liver primary cells), lung cells (human A549, rat lung epithelial cells), mouse fibroblast cells (Balb/3T3), human umbilical vein endothelial cells (HUVECs) and rat neuronal cells (PC12). The in vivo studies were published between 2013 and 2021; four studies were conducted in China, three in Iran, three in Egypt, and two in Saudi Arabia; Bayat et al. explored the protective roles of vitamin A, C and E for ZnONP-induced injuries [10]; Afshari-Kaveh et al. [18] and Moradi et al. [8] evaluated the protective roles of vitamin A, E and A + E for TiO2NP-induced injuries; the other eight studies only assessed the antagonized effects of vitamin E treatment for GNP-, TiO2NP-, ZnONP-, GO (graphene oxide)-, AgNP-induced toxicities [8,9,11,17,25,26,28,29] and one examined the effects of vitamin C for NiNP-induced toxicities [19].

3.3. Quality Assessment

The Toxrtool score was 16 for all studies, indicating that all in vitro studies were of high quality (Table 2). A low risk of bias was considered for the items of performance (random housing), attrition, reporting and other bias in most of animal studies except of the study of Yin et al. [26] (Table 2). Although an unclear risk of bias was assigned for most of the rest items because they were not sufficiently reported, this did not affect the determination of the indicators and the results. Thus, the quality of included in vivo studies was also acceptable.

3.4. Meta-Analysis for In Vitro Studies

3.4.1. Effects of Vitamin E Treatment on Cell Viability

The studies of Wang et al. [12], Yan et al. [31] and Faedmaleki et al. [32] provided the results of cell viability after treatment with six dosages of vitamin E. Cells were exposed to vitamin E for 24 h and 48 h in the study of Wang et al., [12]. Only a 24-h time period was designed for other three studies [27,31,32]. Thus, 25 items of data were used for this meta-analysis. The pooled results showed that vitamin E treatment could significantly improve the cell viability compared with the nanomaterial exposure group (SMD = 4.89; 95%CI, 3.65–6.14; p < 0.001; I2 = 85.2%; p < 0.001) (Table 3; Figure 2). This significant effect was not changed in the subgroup analyses (Table 4).

3.4.2. Effects of Vitamin E Treatment on Cell Apoptosis

Caspase-3 activity was measured to represent cell apoptosis. The study of Wang et al. [12] reported the results of caspase-3 activity after treatment with four dosages (50, 500, 1000, 2000 μM) of vitamin E for 24 h and 48 h. Zhang et al. investigated the effects of vitamin E exposure (27 μM) for 24 h on the caspase-3 activity [27]. Thus, nine data were used for this meta-analysis. The pooled results showed that vitamin E treatment could significantly decrease the caspase-3 activity compared with the nanomaterial exposure group (SMD = −2.07; 95%CI, (−3.25)–(−0.89); p = 0.001; I2 = 80.7%; p < 0.001) (Table 3; Figure 3). The anti-apoptotic ability of vitamin E was only significant after exposure for 48 h with a dosage of ≤ 1000 μM (Table 4).

3.4.3. Effects of Vitamin E Treatment on Oxidative Stress

The effects of vitamin E treatment on ROS (an indicator of oxidative stress) were evaluated in three articles [12,27,31] with ten data because four dosages (50, 500, 1000, 2000 μM) and two treatment durations were included in the study of Wang et al. [12]. The pooled results revealed that vitamin E treatment was associated with reduced ROS levels compared with the nanomaterial exposure group (SMD = −13.07; 95%CI, (−17.85)–(−8.30); p < 0.001; I2 = 90.8%; p < 0.001) (Table 3; Figure 4). Subgroup analyses’ results demonstrated that vitamin E treatment with a dosage of ≤1000 μM significantly inhibited the formation of ROS regardless of nanomaterials types and treatment durations (Table 4).

3.4.4. Effects of Vitamin C Treatment on Cell Viability

Two studies [14,21] with four data (since three concentration gradients were designed for NiONPs in the study of Ahamed et al. [14]) investigated the effects of vitamin C treatment on the cell viability. The meta-analysis results demonstrated that vitamin C intervention could significantly increase the cell viability compared with the nanomaterial exposure group (SMD = 4.19; 95%CI, 2.37–6.01; p < 0.001; I2 = 45.3%; p = 0.140) (Table 3).

3.4.5. Effects of Vitamin C Treatment on Oxidative Stress

Four studies [13,14,21,30] with six data (due to the three concentrations included in the study of Ahamed et al. [14]) investigated the effects of vitamin C treatment on ROS levels. The meta-analysis results demonstrated that vitamin C intervention could significantly decrease the levels of ROS compared with the nanomaterial exposure group (SMD = −6.77; 95%CI, (−12.18)–(−1.36); p = 0.014; I2 = 85.4%; p < 0.001) (Table 3).

3.5. Meta-Analysis for In Vivo studies

3.5.1. Effects of Vitamin E Treatment on Body Weight

Five studies [10,17,18,26,29] with six data (including two ZnONP concentrations included in the study of Baky et al. [17]) monitored the body weight of animals after administration of vitamin E. The meta-analysis results revealed no significant differences in the body weight between vitamin E and nanomaterial exposure groups (p = 0.328) (Table 3). Although the subgroup analysis showed vitamin E could increase the body weight for animals exposed to AgNPs, only one study reported this effect (Table 4) and thus, the conclusion remained indefinite.

3.5.2. Effects of Vitamin E Treatment on Oxidative Stress

Six publications [8,10,11,18,25,28] with seven data (two GO concentrations included in the study of Shang et al. [25]) measured MDA levels; four articles [9,11,25,28] with six data (two concentrations included in the studies of Shang et al. [25] and AL-RASHEED et al. [9]) analyzed GSH levels; three studies [8,10,18] detected TOS, TAC, SOD and GPx; two studies examined OSI [10,18], CAT [8,10], SOD mRNA [10,18], GPx mRNA [10,18] and Nrf2 mRNA expression levels [18,28]. The summary analysis showed that the levels of pro-oxidant indicators [MDA (Figure 5): SMD = −6.37; 95%CI, (−9.11)–(−3.63); p < 0.001; TOS: SMD = −5.89; 95%CI, (−9.94)–(−1.84); p = 0.004; OSI: SMD = -4.19; 95%CI, (−5.73)–(−2.64); p = 0.019] were significantly decreased, while the levels of anti-oxidant indicators (TAC: SMD = 2.48; 95%CI, 1.55–3.41; p < 0.001; SOD: SMD = 4.19; 95%CI, 0.70–7.66; p = 0.019; GPx activity: SMD = 3.99; 95%CI, 2.04–5.93; p < 0.001; GSH: SMD = 5.26; 95%CI, 1.73–8.80; p = 0.004; GPx mRNA expression: SMD = 13.17; 95%CI, 1.21–25.12; p = 0.031) were significantly increased in the vitamin E treatment group relative to the nanomaterial exposure group (Table 3). No significant differences in the CAT activity, the mRNA expression levels of SOD and Nrf2 were present between two groups (p > 0.05) (Table 3). Subgroup analyses demonstrated that only vitamin E treatment for more than two weeks (regardless of dosages) significantly decreased MDA and increased GSH. The improvement effects of vitamin E on GSH may be more sensitive for mice than rats, for ZnONPs, TiO2NPs and GNPs than GO (Table 4).

3.5.3. Effects of Vitamin E Treatment on Inflammation

Five publications [8,9,17,28,29] with seven data measured TNF-α levels; four articles [9,17,28,29] with six data collected IL-6 levels; two studies [8,10,18] with four data assessed CRP levels. The number of real data for analysis of these three inflammatory indicators was larger than the number of articles because two concentrations included in the studies of Baky et al. [17] and AL-RASHEED et al. [9]. Two studies [25,27] with three data investigated the levels of IgE. The mRNA expression level of NF-κB was analyzed in the studies of Moradi et al. [8] and Azim et al. [28]. The summary analysis showed that except of NF-κB, the levels of all other pro-inflammatory indicators were lower in the vitamin E treatment group than those in the nanomaterial exposure group [TNF-α (Figure 6): SMD = −3.29; 95%CI, (−6.24)–(−0.35); p = 0.028; IL-6 (Figure 7): SMD = −13.23; 95%CI, (−17.71)–(−8.76); p < 0.001; CRP: SMD = −5.60; 95%CI, (−6.63)–(−4.57); p < 0.001; IgE: SMD = −4.08; 95%CI, (−5.20)–(−2.95); p < 0.001]. Subgroup analyses indicated that vitamin E treatment only inhibited the production of TNF-α at the early stage (treatment for less than two weeks), but IL-6 at both of the early (≤two weeks) and later (>two weeks) stages (Table 4).

3.5.4. Effects of Vitamin E Treatment on Apoptosis

The pooled analysis of four studies with six data [9,17,26,28] showed that compared with the nanomaterial exposure group, the caspase-3 activity was significantly decreased by vitamin E treatment (SMD = −7.10; 95%CI, (−10.49)–(−3.72); p < 0.001) (Table 3; Figure 8). Subgroup analyses showed that except of AgNPs, vitamin E treatment suppressed apoptosis induced by all other nanomaterials regardless of durations (Table 4).

3.5.5. Effects of Vitamin E Treatment on DNA Damage

Comet assay was performed to evaluate DNA damage for three studies [9,17,28], after which the data about the tail DNA content and the tail length were obtained. The pooled analysis of these three studies with five data revealed a significant decrease in the tail length between two groups (SMD = −7.88; 95%CI, (−11.95)–(−3.81); p < 0.001) (Table 3; Figure 9). There was no significant difference in the tail DNA % (p = 0.283). The improvement effects of vitamin E treatment on the tail length remained significant after subgroup analyses stratified by nanomaterial types and animal model types (Table 4).

3.5.6. Effects of Vitamin E Treatment on Liver Function

Only liver function data (ALT, AST) could be combined for included studies [8,9,28] and thus, a meta-analysis was performed for them. The pooled analysis results showed that the level of ALT (SMD = −7.35; 95%CI, (−11.41)–(−3.29); p < 0.001) was significantly decreased by vitamin E treatment, but not the level of AST (Table 3).

3.5.7. Effects of Vitamin C Treatment on Oxidative Stress

MDA, SOD and CAT were analyzed in two studies to explore the anti-oxidative roles of vitamin C [10,19]. Unexpectedly, the pooled analysis did not detect significant differences in these three indicators between vitamin E and nanomaterial exposure groups (p > 0.05) (Table 3).

3.5.8. Effects of Vitamin A Treatment on Body Weight

Meta-analysis of two studies [10,18] indicated vitamin A treatment could increase the body weight of animals relative to the nanomaterial exposure group (SMD = 2.1; 95%CI, 0.06–4.14; p = 0.043) (Table 3).

3.5.9. Effects of Vitamin A Treatment on Oxidative Stress

Meta-analysis of three studies [8,10,18] indicated vitamin A treatment could reduce the levels of MDA (SMD = −3.17; 95%CI, (−5.50)–(−0.84); p = 0.008) and TOS (SMD = −1.34; 95%CI, (−2.09)–(−0.59); p < 0.001), while increased SOD (SMD = 1.84; 95%CI, 1.01–2.67; p < 0.001) and GPx activity (SMD = 2.73; 95%CI, 1.77–3.7; p < 0.001) (Table 3). Meta-analysis of two studies [8,10] showed the activity of CAT was higher in the vitamin A treatment group relative to the nanomaterial exposure group (SMD = 3.22; 95%CI, 1.04–5.40; p = 0.004) (Table 3).

3.5.10. Effects of Vitamin A + E Treatment on Oxidative Stress

Meta-analysis of two studies [8,18] showed that the level of MDA was reduced in the vitamin A + E treatment group compared with the nanomaterial exposure group (SMD = −8.42; 95%CI, (−11.17)–(−5.67); p = 0.013) (Table 3). TOS, TAC, SOD and GPx were not significantly changed (Table 3).

3.6. Publication Bias and Sensitivity Analysis

Egger’s test showed a publication bias existed in the analysis of several indicators with at least three data analyzed (except of the body weight, p = 0.081; TNF-α, p = 0.251 for in vivo studies with vitamin E treatment) (Table 3). However, significant results were still present for most of indicators [except of SOD (p = 0.435) and GSH (p = 0.198) in in vivo studies with vitamin E treatment, which were no longer significant after being adjusted by the trim and fill method]. Sensitivity analysis results also showed that no individual study affected the synthesized results (Figure 10).

4. Discussion

Although there had meta-analyses to demonstrate that vitamins can exert anti-oxidant and anti-inflammatory activities [15,16,34], no studies investigated their protective roles for nanomaterial-induced injuries until now. In addition, some meta-analysis results found the anti-oxidant and anti-inflammatory functions of vitamins were limited and even indicated vitamins exhibited potential toxic activities [35]. Thus, to prevent the hazard events induced by nanomaterials, but not cause the abuse of health care products, this study included 19 articles and performed a meta-analysis to comprehensively evaluate the roles and mechanisms of vitamins for cells and animals exposed to nanomaterials. Our meta-analysis results showed that vitamin E could antagonize nanomaterial-induced oxidative stress (mainly by reducing ROS, TOS, TAC, OSI, and MDA and increasing GPx), inflammation (significantly reducing the effects on TNF-α, IL-6, CRP, and IgE), improving cytotoxicity (manifested by an increase in the cell viability and a decrease in pro-apoptotic factor caspase-3) and genotoxicity (represented by a reduction in the tail length), which were less changed by subgroup stratifications. Pooled analysis of in vitro studies indicated that vitamin C treatment increased the cell viability and decreased ROS levels, but its anti-oxidant potential was not observed in the meta-analysis of in vivo studies. Vitamin A treatment was shown to decrease MDA (SMD: −3.17 vs. −6.37), TOS (SMD: −1.34 vs. −5.89) and increase GPx (SMD: 2.73 vs. 3.99), but its effects on these indicators seemed to be weaker than vitamin E. Also, the combination of vitamin A with vitamin E seemed not to provide greater anti-oxidant effects than vitamin E (except of MDA that was further reduced by two-fold). Accordingly, we may consider that vitamin E alone supplementation may be more cost-effective to prevent nanomaterial-induced injuries and diseases, especially for populations with occupational exposure.
Based on our results, the preventive roles of vitamin E against nanomaterial-induced injuries (apoptosis and DNA damage) were speculated to be exerted mainly through the following mechanisms: (1) as a fat-soluble vitamin, vitamin E can penetrate the lipid bilayer of the cell membrane and interact with phospholipids to stabilize bilayer structures and decrease the permeability of bilayer membranes [36], which ultimately inhibits the entrance of toxic nanomaterials into human cells [37,38]; (2) vitamin E not only quenches nanomaterial (if they are accidentally permeated to cells)-induced ROS in cell membranes [6,12,31], but also reacts with a lipid hydroperoxyl radical (LOO) by donating hydrogen from its phenolic hydroxyl group at the C-6 position, resulting in the formation of lipid hydroperoxide which was subsequently transformed to non-toxic hydroxide after catalysis by GPx to terminate lipid peroxidation and decrease the levels of the end products of lipid peroxidation (MDA) [39]; (3) previous studies demonstrated that ROS induced an inflammatory response via activation of the mitogen-activated protein kinase-NF-κB signaling pathway [40,41]. Thus, the anti-inflammatory functions of vitamin E may indirectly result from its suppressive effects on oxidative stress. Furthermore, vitamin E was found to directly stimulate the production of cyclic adenosine monophosphate (cAMP) in human peripheral mononuclear cells via the EP2/EP4 receptors and adenylyl cyclase [42], which in turn activated its downstream proteins (protein kinase A and cAMP response element binding) and then suppressed the release of pro-inflammatory cytokines (such as TNF-α and IL-6) from monocytes [43]. Importantly, there was evidence to demonstrate that up-regulation of TNF-α triggered the production of IL-6 [44], while IL-6 stimulated the transcription and synthesis of CRP [45] and IgE [46]. This may be an underlying reason to explain that vitamin E suppressed the levels of TNF-α at the early stage and then IL-6 for a long time as reported in our subgroup analyses.
There are some limitations in this meta-analysis. First, the number of included in vivo and in vitro studies was still limited and the detected indicators were varying in studies, which led to less and no data pooled (such as the anti-inflammatory roles of vitamin C and A; damages on the renal, spleen, heart and brain tissues; the other vitamin types). Second, considerable heterogeneity was present among studies for the analysis of several indicators and the source of heterogeneity could not be removed by the subgroup analysis. Therefore, it is necessary to conduct more experiments on cells, animals, and humans to confirm the conclusions of our study.

5. Conclusions

This meta-analysis of 19 in vitro and in vivo studies provides evidence that supplementation with vitamins (especially vitamin E) may be beneficial to prevent nanomaterial-induced cytotoxicity and genotoxicity by exerting anti-oxidative and anti-inflammatory activities. Our findings support the clinical recommendations of vitamin E intake for workers with occupational exposure to nanomaterials. However, our conclusions are still needed to be confirmed by analysis of more studies of high-quality and lack of heterogeneity.

Author Contributions

D.X.: conceptualization, methodology, data curation, formal analysis, writing—original draft. J.H.: methodology, investigation, software, formal analysis. Z.Y.: visualization, resources. T.W.: investigation, visualization. W.X.: investigation, visualization; Q.M.: investigation, visualization; K.C.: investigation, funding acquisition. X.L.: conceptualization, supervision, funding acquisition, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

Financial support from National Key Research and Development Program of China (2017YFA0204600) and Project funded by China Postdoctoral Science Foundation (2017M621322, 2018T110324) is greatly acknowledged.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data can be obtained in published articles.

Conflicts of Interest

All authors declare that they have no competing interest. Tong Wu is an employee of Shanghai Jing Rui Yang Industrial Co., Ltd. Wei Xu is an employee of Shanghai Nutri-woods Bio-Technology Co., Ltd. Qingyang Meng is an employee of Shanghai Pechoin Daily Chemical Co., Ltd. Also, no any hidden conflict of interest exists by these companies.

References

  1. Mabrouk, M.; Das, D.B.; Salem, Z.A.; Beherei, H.H. Nanomaterials for biomedical applications: Production, characterisations, recent trends and difficulties. Molecules 2021, 26, 1077. [Google Scholar] [CrossRef] [PubMed]
  2. Ranjha, M.; Shafique, B.; Rehman, A.; Mehmood, A.; Ali, A.; Zahra, S.M.; Roobab, U.; Singh, A.; Ibrahim, S.A.; Siddiqui, S.A. Biocompatible nanomaterials in food science, technology, and nutrient drug delivery: Recent developments and applications. Front. Nutr. 2021, 8, 778155. [Google Scholar] [CrossRef] [PubMed]
  3. Fytianos, G.; Rahdar, A. Nanomaterials in cosmetics: Recent updates. Nanomaterials 2020, 10, 979. [Google Scholar] [CrossRef] [PubMed]
  4. De Matteis, V. Exposure to inorganic nanoparticles: Routes of entry, immune response, biodistribution and In Vitro/In Vivo toxicity evaluation. Toxics 2017, 5, 29. [Google Scholar] [CrossRef] [Green Version]
  5. Labouta, H.I.; Asgarian, N.; Rinker, K.; Cramb, D.T. Meta-analysis of nanoparticle cytotoxicity via data-mining the literature. ACS Nano 2019, 13, 1583–1594. [Google Scholar] [CrossRef]
  6. An, H.; Ling, C.; Xu, M.; Hu, M.; Wang, H.; Liu, J.; Song, G. Oxidative damage induced by nano-titanium dioxide in rats and mice: A systematic review and meta-analysis. Biol. Trace Elem. Res. 2020, 194, 184–202. [Google Scholar] [CrossRef]
  7. Vilas-Boas, V.; Vinken, M. Hepatotoxicity induced by nanomaterials: Mechanisms and In Vitro models. Arch. Toxicol. 2021, 95, 27–52. [Google Scholar] [CrossRef]
  8. Moradi, A.; Ziamajidi, N.; Ghafourikhosroshahi, A.; Abbasalipourkabir, R. Effects of vitamin A and vitamin E on attenuation of titanium dioxide nanoparticles-induced toxicity in the liver of male Wistar rats. Mol. Biol. Rep. 2019, 46, 2919–2932. [Google Scholar] [CrossRef]
  9. Al-Rasheed, N.M.; Baky, N.A.; Faddah, L.M.; Fatani, A.J.; Hasan, I.H.; Mohamad, R.A. Prophylactic role of α-lipoic acid and vitamin E against zinc oxide nanoparticles induced metabolic and immune disorders in rat’s liver. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 1813–1828. [Google Scholar]
  10. Bayat, M.; Daei, S.; Ziamajidi, N.; Abbasalipourkabir, R.; Nourian, A. The protective effects of vitamins A, C, and E on zinc oxide nanoparticles (ZnO NPs)-induced liver oxidative stress in male Wistar rats, drug and chemical toxicology. Drug Chem. Toxicol. 2021, 1–10. [Google Scholar] [CrossRef]
  11. Abdelhalim, M.A.K.; Qaid, H.A.; Al-Mohy, Y.H.; Ghannam, M.M. The protective roles of vitamin E and α-lipoic acid against nephrotoxicity, lipid peroxidation, and inflammatory damage induced by gold nanoparticles. Int. J. Nanomed. 2020, 15, 729–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Wang, J.; Sun, P.; Bao, Y.; Dou, B.; Song, D.; Li, Y. Vitamin E renders protection to PC12 cells against oxidative damage and apoptosis induced by single-walled carbon nanotubes. Toxicol. Vitr. 2012, 26, 32–41. [Google Scholar] [CrossRef] [PubMed]
  13. Sharma, C.S.; Sarkar, S.; Periyakaruppan, A.; Barr, J.; Wise, K.; Thomas, R.; Wilson, B.L.; Ramesh, G.T. Single-walled carbon nanotubes induces oxidative stress in rat lung epithelial cells. J. Nanosci. Nanotechnol. 2007, 7, 2466–2472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ahamed, M.; Ali, D.; Alhadlaq, H.A.; Akhtar, M.J. Nickel oxide nanoparticles exert cytotoxicity via oxidative stress and induce apoptotic response in human liver cells (HepG2). Chemosphere 2013, 93, 2514–2522. [Google Scholar] [CrossRef] [PubMed]
  15. Nguyen, Y.; Sigaux, J.; Letarouilly, J.G. Efficacy of oral vitamin supplementation in inflammatory rheumatic disorders: A systematic review and meta-analysis of randomized controlled trials. Nutrients 2020, 13, 107. [Google Scholar] [CrossRef]
  16. Mansournia, M.A.; Ostadmohammadi, V.; Doosti-Irani, A.; Ghayour-Mobarhan, M.; Ferns, G.; Akbari, H.; Ghaderi, A.; Talari, H.R.; Asemi, Z. The effects of vitamin D supplementation on biomarkers of inflammation and oxidative stress in diabetic patients: A systematic review and meta-analysis of randomized controlled trials. Horm. Metab. Res. 2018, 50, 429–440. [Google Scholar] [CrossRef] [Green Version]
  17. Baky, N.A.; Faddah, L.M.; Al-Rasheed, N.M.; Al-Rasheed, N.M.; Fatani, A.J. Induction of inflammation, DNA damage and apoptosis in rat heart after oral exposure to zinc oxide nanoparticles and the cardioprotective role of α-lipoic acid and vitamin E. Drug Res. 2013, 63, 228–236. [Google Scholar] [CrossRef]
  18. Afshari-Kaveh, M.; Abbasalipourkabir, R.; Nourian, A.; Ziamajidi, N. The protective effects of vitamins A and E on titanium dioxide nanoparticles (nTiO2)-induced oxidative stress in the spleen tissues of male wistar rats. Biol. Trace Elem. Res. 2021, 199, 3677–3687. [Google Scholar] [CrossRef]
  19. Kong, L.; Hu, W.; Lu, C.; Cheng, K.; Tang, M. Mechanisms underlying nickel nanoparticle induced reproductive toxicity and chemo-protective effects of vitamin C in male rats. Chemosphere 2019, 218, 259–265. [Google Scholar] [CrossRef]
  20. Schneider, K.; Schwarz, M.; Burkholder, I.; Kopp-Schneider, A.; Edler, L.; Kinsner-Ovaskainen, A.; Hartung, T.; Hoffmann, S. “ToxRTool”, a new tool to assess the reliability of toxicological data. Toxicol. Lett. 2009, 189, 138–144. [Google Scholar] [CrossRef]
  21. Sanches, P.L.; Geaquinto, L.R.O.; Cruz, R.; Schuck, D.C.; Lorencini, M.; Granjeiro, J.M.; Ribeiro, A.R.L. Toxicity evaluation of TiO(2) nanoparticles on the 3D skin model: A systematic review. Front. Bioeng. Biotechnol. 2020, 8, 575. [Google Scholar] [CrossRef] [PubMed]
  22. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hafez, A.A.; Naserzadeh, P.; Ashtari, K.; Mortazavian, A.M.; Salimi, A. Protection of manganese oxide nanoparticles-induced liver and kidney damage by vitamin D. Regul. Toxicol. Pharmacol. 2018, 98, 240–244. [Google Scholar] [CrossRef]
  24. Naskhi, A.; Jabbari, S.; Othman, G.Q.; Aziz, F.M.; Salihi, A. Vitamin K1 as a potential molecule for reducing single-walled carbon nanotubes-stimulated α-synuclein structural changes and cytotoxicity. Int. J. Nanomed. 2019, 14, 8433–8444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Shang, S.; Li, J.; Zhao, Y.; Xi, Z.; Lu, Z.; Li, B.; Yang, X.; Li, R. Oxidized graphene-aggravated allergic asthma is antagonized by antioxidant vitamin E in Balb/c mice. Environ. Sci. Pollut. Res. Int. 2017, 24, 1784–1793. [Google Scholar] [CrossRef]
  26. Yin, N.; Yao, X.; Zhou, Q.; Faiola, F.; Jiang, G. Vitamin E attenuates silver nanoparticle-induced effects on body weight and neurotoxicity in rats. Biochem. Biophys. Res. Commun. 2015, 458, 405–410. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, Q.; Liu, Z.; Du, J.; Qin, W.; Lu, M.; Cui, H.; Li, X.; Ding, S.; Li, R.; Yuan, J. Dermal exposure to nano-TiO2 induced cardiovascular toxicity through oxidative stress, inflammation and apoptosis. J. Toxicol. Sci. 2019, 44, 35–45. [Google Scholar] [CrossRef]
  28. Azim, S.A.; Darwish, H.A.; Rizk, M.Z.; Ali, S.A.; Kadry, M.O. Amelioration of titanium dioxide nanoparticles-induced liver injury in mice: Possible role of some antioxidants. Exp. Toxicol. Pathol. 2015, 67, 305–314. [Google Scholar] [CrossRef]
  29. Abdelkarem, H.M.; Fadda, L.H.; El-Sayed, E.M.; Radwan, O.K. Potential role of l-arginine and vitamin E against bone loss induced by nano-zinc oxide in rats. J. Diet. Suppl. 2018, 15, 300–310. [Google Scholar] [CrossRef]
  30. Fukui, H.; Iwahashi, H.; Nishio, K.; Hagihara, Y.; Yoshida, Y.; Horie, M. Ascorbic acid prevents zinc oxide nanoparticle-induced intracellular oxidative stress and inflammatory responses. Toxicol. Ind. Health 2017, 33, 687–695. [Google Scholar] [CrossRef]
  31. Yan, X.; Liu, Y.; Xie, T.; Liu, F. α-Tocopherol protected against cobalt nanoparticles and cocl2 induced cytotoxicity and inflammation in Balb/3T3 cells. Immunopharmacol. Immunotoxicol. 2018, 40, 179–185. [Google Scholar] [CrossRef] [PubMed]
  32. Faedmaleki, F.; Shirazi, F.H.; Ejtemaeimehr, S.; Anjarani, S.; Salarian, A.A.; Ashtiani, H.A.; Rastegar, H. Study of silymarin and vitamin E protective effects on silver nanoparticle toxicity on mice liver primary cell culture. Acta Med. Iran. 2016, 54, 85–95. [Google Scholar] [PubMed]
  33. Liu, Y.; Hong, H.; Lu, X.; Wang, W.; Liu, F.; Yang, H. L-ascorbic acid protected against extrinsic and intrinsic apoptosis induced by cobalt nanoparticles through ROS attenuation. Biol. Trace Elem. Res. 2017, 175, 428–439. [Google Scholar] [CrossRef]
  34. Violi, F.; Nocella, C.; Loffredo, L.; Carnevale, R.; Pignatelli, P. Interventional study with vitamin E in cardiovascular disease and meta-analysis. Free Radic. Biol. Med. 2022, 178, 26–41. [Google Scholar] [CrossRef]
  35. Rai, S.N.; Singh, P.; Steinbusch, H.W.M. The role of vitamins in neurodegenerative disease: An update. Biomedicines 2021, 9, 1284. [Google Scholar] [CrossRef] [PubMed]
  36. Quinn, P.J. Molecular associations of vitamin E. Vitam. Horm. 2007, 76, 67–98. [Google Scholar]
  37. Ziglari, T.; Anderson, D.S. Determination of the relative contribution of the non-dissolved fraction of ZnO NP on membrane permeability and cytotoxicity. Inhal. Toxicol. 2020, 32, 86–95. [Google Scholar] [CrossRef] [PubMed]
  38. Hussain, S. Measurement of nanoparticle-induced mitochondrial membrane potential alterations. Methods Mol. Biol. 2019, 1894, 123–131. [Google Scholar]
  39. Traber, M.G.; Stevens, J.F. Vitamins C and E: Beneficial effects from a mechanistic perspective. Free Radic. Biol. Med. 2011, 51, 1000–1013. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, Z.; Yao, X.; Jiang, W.; Li, W.; Zhu, S.; Liao, C.; Zou, L.; Ding, R.; Chen, J. Advanced oxidation protein products induce microglia-mediated neuroinflammation via MAPKs-NF-κB signaling pathway and pyroptosis after secondary spinal cord injury. J. Neuroinflamm. 2020, 17, 90. [Google Scholar] [CrossRef]
  41. Wu, L.; Pan, Y. Reactive oxygen species mediate TNF-α-induced inflammatory response in bone marrow mesenchymal cells. Iran. J. Basic Med. Sci. 2019, 22, 1296–1301. [Google Scholar] [PubMed]
  42. Salinthone, S.; Kerns, A.R.; Tsang, V.; Carr, D.W. α-Tocopherol (vitamin E) stimulates cyclic AMP production in human peripheral mononuclear cells and alters immune function. Mol. Immunol. 2013, 53, 173–178. [Google Scholar] [CrossRef] [PubMed]
  43. Qi, Y.C.; Duan, G.Z.; Mao, W.; Liu, Q.; Zhang, Y.L.; Li, P.F. Taurochenodeoxycholic acid mediates cAMP-PKA-CREB signaling pathway. Chin. J. Nat. Med. 2020, 18, 898–906. [Google Scholar] [CrossRef]
  44. Chen, M.; Chen, Z.; Huang, D.; Sun, C.; Xie, J.; Chen, T.; Zhao, X.; Huang, Y.; Li, D.; Wu, B.; et al. Myricetin inhibits TNF-α-induced inflammation in A549 cells via the SIRT1/NF-κB pathway. Pulm. Pharmacol. Ther. 2020, 65, 102000. [Google Scholar] [CrossRef] [PubMed]
  45. Srivastava, R.A.K.; Cornicelli, J.A.; Markham, B.; Bisgaier, C.L. Gemcabene, a first-in-class lipid-lowering agent in late-stage development, down-regulates acute-phase C-reactive protein via C/EBP-δ-mediated transcriptional mechanism. Mol. Cell Biochem. 2018, 449, 167–183. [Google Scholar] [CrossRef] [Green Version]
  46. Scheid, C.; Young, R.; McDermott, R.; Fitzsimmons, L.; Scarffe, J.H.; Stern, P.L. Immune function of patients receiving recombinant human interleukin-6 (IL-6) in a phase I clinical study: Induction of C-reactive protein and IgE and inhibition of natural killer and lymphokine-activated killer cell activity. Cancer Immunol. Immunother. 1994, 38, 119–126. [Google Scholar]
Figure 1. PRISMA flowchart.
Figure 1. PRISMA flowchart.
Nutrients 14 02214 g001
Figure 2. Forest plots showing the protective effects of vitamin E on the cell viability compared with the nanomaterial exposure group. a, b, c, d, e, f of the study of Wang et al. represent treatment with 10, 50, 200, 500, 1000, 2000 μM of vitamin E for 24 h; g, h, I, j, k, l of the study of Wang et al. represent treatment with 10, 50, 200, 500, 1000, 2000 μM of vitamin E for 48 h. a, b, c, d, e, f of the study of Faedmaleki et al. represent treatment with 50, 250, 500, 1000, 2500, 5000 μM of vitamin E for 24 h. a, b, c, d, e, f of the study of Yan et al. represent treatment with 2, 5, 10, 25, 50, 100 μM of vitamin E for 24 h. SMD, standardized mean difference; CI, confidence interval [12,27,31,32].
Figure 2. Forest plots showing the protective effects of vitamin E on the cell viability compared with the nanomaterial exposure group. a, b, c, d, e, f of the study of Wang et al. represent treatment with 10, 50, 200, 500, 1000, 2000 μM of vitamin E for 24 h; g, h, I, j, k, l of the study of Wang et al. represent treatment with 10, 50, 200, 500, 1000, 2000 μM of vitamin E for 48 h. a, b, c, d, e, f of the study of Faedmaleki et al. represent treatment with 50, 250, 500, 1000, 2500, 5000 μM of vitamin E for 24 h. a, b, c, d, e, f of the study of Yan et al. represent treatment with 2, 5, 10, 25, 50, 100 μM of vitamin E for 24 h. SMD, standardized mean difference; CI, confidence interval [12,27,31,32].
Nutrients 14 02214 g002
Figure 3. Forest plots showing the protective effects of vitamin E on Caspase-3 levels of cells compared with the nanomaterial exposure group. a, b, c, d of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 24 h; e, f, g, h of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 48 h. SMD, standardized mean difference; CI, confidence interval [12,27].
Figure 3. Forest plots showing the protective effects of vitamin E on Caspase-3 levels of cells compared with the nanomaterial exposure group. a, b, c, d of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 24 h; e, f, g, h of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 48 h. SMD, standardized mean difference; CI, confidence interval [12,27].
Nutrients 14 02214 g003
Figure 4. Forest plots showing the protective effects of vitamin E on ROS levels of cells compared with the nanomaterial exposure group. a, b, c, d of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 24 h; e, f, g, h of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 48 h. ROS, reactive oxygen species; SMD, standardized mean difference; CI, confidence interval [12,27,31].
Figure 4. Forest plots showing the protective effects of vitamin E on ROS levels of cells compared with the nanomaterial exposure group. a, b, c, d of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 24 h; e, f, g, h of the study of Wang et al. represent treatment with 50, 500, 1000, 2000 μM of vitamin E for 48 h. ROS, reactive oxygen species; SMD, standardized mean difference; CI, confidence interval [12,27,31].
Nutrients 14 02214 g004
Figure 5. Forest plots showing the protective effects of vitamin E on MDA levels of murine models compared with the nanomaterial exposure group. a, b of the study of Shang et al. represent treatment with 0.4 and 4 mg/kg of graphene oxide. MDA, malonaldehyde; SMD, standardized mean difference; CI, confidence interval [8,10,11,18,25,28].
Figure 5. Forest plots showing the protective effects of vitamin E on MDA levels of murine models compared with the nanomaterial exposure group. a, b of the study of Shang et al. represent treatment with 0.4 and 4 mg/kg of graphene oxide. MDA, malonaldehyde; SMD, standardized mean difference; CI, confidence interval [8,10,11,18,25,28].
Nutrients 14 02214 g005
Figure 6. Forest plots showing the protective effects of vitamin E on TNF-α levels of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. TNF, tumor necrosis factor; SMD, standardized mean difference; CI, confidence interval [8,9,17,28,29].
Figure 6. Forest plots showing the protective effects of vitamin E on TNF-α levels of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. TNF, tumor necrosis factor; SMD, standardized mean difference; CI, confidence interval [8,9,17,28,29].
Nutrients 14 02214 g006
Figure 7. Forest plots showing the protective effects of vitamin E on IL-6 levels of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. IL, interleukin; SMD, standardized mean difference; CI, confidence interval [9,17,28,29].
Figure 7. Forest plots showing the protective effects of vitamin E on IL-6 levels of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. IL, interleukin; SMD, standardized mean difference; CI, confidence interval [9,17,28,29].
Nutrients 14 02214 g007
Figure 8. Forest plots showing the protective effects of vitamin E on Caspase-3 activity of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. SMD, standardized mean difference; CI, confidence interval [9,17,26,28].
Figure 8. Forest plots showing the protective effects of vitamin E on Caspase-3 activity of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. SMD, standardized mean difference; CI, confidence interval [9,17,26,28].
Nutrients 14 02214 g008
Figure 9. Forest plots showing the protective effects of vitamin E on tail length of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. SMD, standardized mean difference; CI, confidence interval [9,17,28].
Figure 9. Forest plots showing the protective effects of vitamin E on tail length of murine models compared with the nanomaterial exposure group. a, b of the study of Baky et al. and AL-RASHEED et al. represent treatment with 600 and 1000 mg/kg of ZnONPs. SMD, standardized mean difference; CI, confidence interval [9,17,28].
Nutrients 14 02214 g009
Figure 10. Sensitivity analysis for MDA levels of murine models treated with vitamin E. MDA, malonaldehyde; CI, confidence interval [8,10,11,18,25,28].
Figure 10. Sensitivity analysis for MDA levels of murine models treated with vitamin E. MDA, malonaldehyde; CI, confidence interval [8,10,11,18,25,28].
Nutrients 14 02214 g010
Table 1. Basic characteristics of included articles.
Table 1. Basic characteristics of included articles.
AuthorYearCountryNanomaterial TypeNanomaterial DoseVitamin
Type
Cell (Animal) TypeVitamin DoseVitamin Treatment DurationNo.Outcomes
In vitroZhang Q [27]2019ChinaTiO2NPs56.25 μg/mLVEHUVECs27 μM24 h14Cell viability, caspase 3, ROS
Yan X [31]2018ChinaCoNPs100 μMVEBalb/3T3 cells2, 5, 10, 25, 50, 100 μM24 h6Cell viability, ROS
Faedmaleki F [32]2016IranAgNPs121.7 μg/mLVEMice liver primary cells 50, 250, 500, 1000, 2500, 5000 μM24 h12Cell viability
Liu Y [33]2016ChinaCoNPs50 μMVCBalb/3T3 cells50 μM24 h6Cell viability, ROS
Fukui H [30]2016JapanZnONPs0.1 mg/mLVCA549 cells5000 μM6 h6ROS
Ahamed M [14]2013Saudi ArabiaNiONPs25 μg/mLVCHepG2 cells1500 μM24 h6Cell viability, ROS
Wang J [12]2012ChinaSWCNTs50 μg/mLVEPC12 cells10, 50, 200, 500, 1000, 2000 μM24, 48 h12Cell viability, caspase 3, ROS
Sharma CS [13]2007USASWCNTs10 μg/mLVCRat lung epithelial cells1000 μM6 h16ROS
In vivoBayat M [10]2021IranZnONPs200 mg/kgVERats100 IU/kg3 weeks12Body weight, MDA, TOS, TAC, OSI, SOD, GPx, CAT
Bayat M [10]2021IranZnONPs200 mg/kgVCRats100 IU/kg3 weeks12MDA, SOD, CAT
Bayat M [10]2021IranZnONPs200 mg/kgVARats100 IU/kg3 weeks12Body weight, MDA, TOS, TAC, OSI, SOD, GPx, CAT
Afshari-Kaveh M [18]2020IranTiO2NPs300 mg/kgVERats100 IU/kg3 weeks12Body weight, MDA, TOS, TAC, OSI, SOD, GPx, Nrf2
Afshari-Kaveh M [18]2020IranTiO2NPs300 mg/kgVARats100 IU/kg3 weeks12Body weight, MDA, TOS, TAC, OSI, SOD, GPx,
Afshari-Kaveh M [18]2020IranTiO2NPs300 mg/kgVA + VERats100 IU/kg3 weeks12MDA, TOS, TAC, SOD, GPx
Abdelhalim MAK [11]2020Saudi ArabiaGNPs50 µLVERats200 mg/kg1 week12MDA, GSH
Moradi A [8]2019IranTiO2NPs300 mg/kgVERats100 IU/kg2 weeks12MDA, TOS, TAC, SOD, GPx, CAT, NF-κB, TNF-α, ALT, AST
Moradi A [8]2019IranTiO2NPs300 mg/kgVARats100 IU/kg2 weeks12MDA, TOS, TAC, SOD, GPx, CAT
Moradi A [8]2019IranTiO2NPs300 mg/kgVA + VERats100 IU/kg2 weeks12MDA, TOS, TAC, SOD, GPx
Zhang Q [27]2019ChinaTiO2NPs500 mg/kgVEMice100 mg/kg6 weeks14IgE
Kong L [19]2019ChinaNiNPs45 mg/kgVCRats1 g/L2 weeks20MDA, SOD, CAT
Abdelkarem HM [29]2018EgyptZnONPs600 mg/kgVERats200 mg/kg3 weeks20Body weight, TNF-α, IL-6
Shang S [25]2016ChinaGO0.4, 4 mg/kgVEMice100 mg/kg4.6 weeks14MDA, GSH, ROS, IgE
Yin N [26]2015ChinaAgNPs2 mg/kgVERats100 mg/kg4.6 weeks6Body weight, caspase-3
Azim SA [28]2015EgyptTiO2NPs150 mg/kgVEMice100 mg/kg2 weeks20MDA, GSH, NF-κB, Nrf2, caspase-3, tail length, DNA%,TNF-α, IL-6, ALT, AST
AL-RASHEED NM [9]2014Saudi ArabiaZnONPs600,1000 mg/kgVERats100 mg/kg3 weeks20GSH, caspase-3, tail length, DNA%,TNF-α, IL-6, CRP, ALT
Baky NA [17]2013EgyptZnONPs600, 1000 mg/kgVERats100 mg/kg3 weeks20Body weight, caspase-3, tail length, DNA%, TNF-α, IL-6, CRP
TiO2NPs, titanium dioxide nanoparticles; ZnONPs, zinc oxide nanoparticles; CoNPs, cobalt nanoparticles; GNPs, gold nanoparticles; NiONPs, nickel oxide nanoparticles; AgNPs, silver nanoparticles; NiNPs, nickel nanoparticles; GO, graphene oxide; SWCNTs, single walled carbon nanotubes; VE, vitamin E; VA, vitamin A; VC, vitamin C; HUVECs, human umbilical vein endothelial cells; ROS, reactive oxygen species; MDA, malonaldehyde; TOS, total oxidant status; TAC, total antioxidant capacity; OSI, oxidative stress index; SOD, superoxide dismutase; GSH, glutathione; GPx, glutathione peroxidase; CAT, catalase; TNF, tumor necrosis factor; NF-κB, nuclear factor kappaB; IL, interleukin; CRP, C-reactive protein; Nrf2, nuclear factor E2-related factor 2; AST, aspartate aminotransferase; ALT, alanine aminotransferase.
Table 2. Quality assessments.
Table 2. Quality assessments.
Toxrtool Checklist of Study Quality
Reference
(In Vitro)
(1)(2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)TotalReliability of Evidence
Zhang Q [27]10101111111111111116Reliable without restrictions
Yan X [31]10101111111111111116Reliable without restrictions
Faedmaleki F [32]10101111111111111116Reliable without restrictions
Liu Y [33]10101111111111111116Reliable without restrictions
Fukui H [30]10101111111111111116Reliable without restrictions
Ahamed M [14]10101111111111111116Reliable without restrictions
Wang J [12]10101111111111111116Reliable without restrictions
Sharma CS [13]10101111111111111116Reliable without restrictions
SYRCLE Checklist of Study Quality
Reference
(In Vivo)
Selection BiasPerformance BiasDetection BiasAttrition BiasReporting BiasOther
SGBCACRHBIROABOAIODSOR
Bayat M [10]UnclearLowUnclearLowUnclearUnclearUnclearLowLowLow
Afshari-Kaveh M [18]UnclearLowUnclearLowUnclearLowUnclearLowLowLow
Abdelhalim MAK [11]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Moradi A [8]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Zhang Q [27]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Kong L [19]UnclearUnclearUnclearLowUnclearLowUnclearLowLowLow
Abdelkarem HM [29]UnclearLowUnclearLowUnclearUnclearUnclearLowLowLow
Shang S [25]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Yin N [26]UnclearUnclearUnclearUnclearUnclearUnclearUnclearLowLowLow
Azim SA [28]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Al-Rasheed NM [9]UnclearUnclearUnclearLowUnclearUnclearUnclearLowLowLow
Baky NA [17]UnclearLowUnclearLowUnclearUnclearUnclearLowLowLow
(1) Test substance identification; (2) substance purity statement; (3) the source/origin information of the substance; (4) information on physicochemical properties of the test item given; (5) cell culture description; (6) the source/origin of cell culture; (7) necessary information on cell culture properties, conditions of cultivation and maintenance; (8) the method of vitamin administration; (9) doses or concentration statement; (10) frequency and duration of exposure as well as time-points of observations statement; (11) have negative controls; (12) have positive controls; (13) the number of replicates; (14) are the study endpoint(s) and their method(s) of determination clearly described?; (15) is the description of the study results for all endpoints investigated transparent and complete?; (16) are the statistical methods for data analysis given and applied in a transparent manner?; (17) is the study design chosen appropriate for obtaining the substance-specific data aimed at?; (18) are the quantitative study results reliable? Each item of Toxrtool obtained a score of 1 if the article satisfied the criteria; 1; otherwise, the score of 0 was given for the articles. SG, sequence generation; BC, baseline characteristics; AC, allocation concealment; RH, random housing; BI, blinding of investigators; ROA, random outcome assessment; BOA, blinding of outcome assessor; IOD, incomplete outcome data; SOR, selective outcome reporting.
Table 3. Meta-analysis results.
Table 3. Meta-analysis results.
StudiesNo.SMD95%CIpE-ValueI2pH-ValueModelEgger
VE (in vitro)
  Cell viability254.893.65, 6.14<0.00185.2<0.001R<0.001
  Caspase-3 9−2.07−3.25, −0.890.00180.7<0.001R<0.001
  ROS 10−13.07−17.85, −8.30<0.00190.8<0.001R<0.001
VC (in vitro)
  Cell viability44.192.37, 6.01<0.00145.30.140F<0.001
  ROS6−6.77−12.18, −1.360.01485.4<0.001R0.002
VE (in vivo)
  Body weight60.52−0.52, 1.560.32878.8<0.001R0.081
  MDA7−6.37−9.11, −3.63<0.00182.9<0.001R<0.001
  TOS3−5.89−9.94, −1.840.00484.10.002R0.006
  TAC32.481.55, 3.41<0.00134.50.217F0.035
  OSI2−4.19−5.73, −2.64<0.0019.20.294F-
  SOD activity34.190.70, 7.660.01986.00.001R0.016
  GPx activity33.992.04, 5.93<0.00158.80.088R0.023
  CAT activity217.33−9.19, 43.850.20092.9<0.001R-
  GSH65.261.73, 8.800.00493.30.000R0.004
  SOD mRNA27.52−2.96, 18.000.15991.50.001R-
  GPx mRNA213.171.21, 25.120.03184.30.012R-
  Nrf2 mRNA2−27.48−88.49, 33.540.37797.4<0.001R-
  TNF-α7−3.29−6.24, −0.350.02892.7<0.001R0.251
  IL-66−13.23−17.71, −8.76<0.00186.1<0.001R<0.001
  CRP4−5.60−6.63, −4.57<0.00130.70.228F<0.001
  IgE3−4.08−5.20, −2.95<0.0010.00.631F0.009
  NF-κB mRNA2−26.00−57.03, 5.020.10094.4<0.001R-
  Caspase-36−7.10−10.49, −3.72<0.00192.2<0.001R0.019
  Tail length5−7.88−11.95, −3.81<0.00195.6<0.001R0.001
  Tail DNA %5−0.94−2.66, 0.780.28390.5<0.001R0.006
  ALT4−7.35−11.41, −3.29<0.00190.7<0.001R0.021
  AST2−14.61−35.90, 6.690.17995.8<0.001R-
VC (in vivo)
  MDA2−1.49−4.13, 1.150.26886.70.006R-
  SOD22.70−4.04, 9.430.43394.7<0.001R-
  CAT210.17−10.17, 30.500.32794.7<0.001R-
VA (in vivo)
  Body weight22.100.06, 4.140.04371.10.063R-
  MDA3−3.17−5.50, −0.840.00878.60.009R0.050
  TOS3−1.34−2.09, −0.59<0.00116.60.302F<0.001
  TAC31.75−0.25, 3.750.08681.80.004R0.038
  SOD31.841.01, 2.67<0.00142.20.177F<0.001
  GPx32.731.77, 3.70<0.0010.00.441F0.008
  OSI2−3.41−8.63, 1.800.20090.60.001R-
  CAT23.221.04, 5.400.00461.60.107R-
  SOD mRNA29.75−4.80, 24.310.18992.4<0.001R-
  GPx mRNA213.32−4.24, 30.870.13791.20.001R-
VA + VE (in vivo)
  MDA2−8.42−11.17, −5.670.0130.00.461F-
  TOS2−3.90−10.54, 2.750.25092.4<0.001R-
  TAC22.00−0.01, 4.010.05171.40.062R-
  SOD22.45−0.29, 5.200.08080.60.023R-
  GPx23.67−0.79, 8.120.10787.20.005R-
ROS, reactive oxygen species; MDA, malonaldehyde; TOS, total oxidant status; TAC, total antioxidant capacity; OSI, oxidative stress index; SOD, superoxide dismutase; GSH, glutathione; GPx, glutathione peroxidase; CAT, catalase; TNF, tumor necrosis factor; NF-κB, nuclear factor kappaB; IL, interleukin; CRP, C-reactive protein; Nrf2, nuclear factor E2-related factor 2; AST, aspartate aminotransferase; ALT, alanine aminotransferase; SMD, standardized mean difference; CI, confidence interval; F, fixed-effects; R, random-effects; pH-value, significance for heterogeneity; pE-value, significance for treatment effects. Bold indicated the outcomes significantly changed by vitamins.
Table 4. Subgroup analysis.
Table 4. Subgroup analysis.
Studies No.SMD95%CIpE-ValueI2pH-ValueModel
VE (in vitro)
Cell viability
 Nanomaterials typeTiO2NPs130.7818.42, 43.14<0.001--R
SWCNTs123.892.48, 5.30<0.00183.8<0.001R
AgNPs611.035.23, 16.83<0.00191.8<0.001R
CoNPs62.641.15, 4.12<0.00146.60.096R
 Duration24 h194.683.24, 6.13<0.00185.1<0.001R
48 h65.632.73, 8.52<0.00187.2<0.001R
 Dosage≤1000 μM214.152.92, 5.38<0.00183.8<0.001R
>1000 μM49.184.82, 13.53<0.00177.20.004R
Caspase-3
 Nanomaterials typeTiO2NPs1−20.37−28.58, −12.15<0.001--R
SWCNTs8−1.58−2.43, −0.72<0.00166.20.004R
 Duration24 h5−1.51−3.12, 0.100.06682.9<0.001R
48 h4−2.78−4.53, −1.030.00275.90.006R
 Dosage≤1000 μM7−1.73−2.97, −0.460.00879.9<0.001R
>1000 μM2−3.85−9.10, 1.390.15089.50.002R
ROS
 Nanomaterials typeTiO2NPs1−4.08−6.02, −2.14<0.001--R
SWCNTs8−17.07−24.40, −9.75<0.00192.6<0.001R
CoNPs1−13.91−17.85, −8.300.005--R
 Duration24 h6−8.02−11.75, −4.30<0.00186.7<0.001R
48 h4−46.94−76.37, −17.510.00290.7<0.001R
 Dosage≤1000 μM8−10.95−15.34, −6.55<0.00189.6<0.001R
>1000 μM2−91.15−242.86, 60.570.23993.7<0.001R
VE (in vivo)
Body weight
 Nanomaterials typeZnONPs40.07−1.08, 1.230.90180.70.001R
TiO2NPs11.04−0.18, 2.260.095--R
AgNPs13.040.40, 5.690.024--R
 Dosage≤100 mg/kg50.81−0.48, 2.110.22081.0<0.001R
>100 mg/kg10.52−0.51, −1.400.267--R
MDA
 Nanomaterials typeZnONPs1−5.14−7.66, −2.62<0.001--R
TiO2NPs3−17.00−28.58, −5.420.00493.3<0.001R
GNPs1−4.03−6.12, −1.93<0.001--R
GO2−5.31−6.99, −3.63<0.0010.00.688F
 Dosage≤100 mg/kg6−7.11−10.50, −3.71<0.00184.8<0.001R
>100 mg/kg1−4.03−6.12, −1.93<0.001--R
 Duration≤2 weeks2−41.95−113.95, −30.050.25396.7<0.001R
>2 weeks5−5.15−6.26, −4.05<0.0010.00.509F
 Sample sourceLiver3−14.14−24.25, −4.030.00693.6<0.001R
Spleen1−7.72−11.29, −4.16<0.001--R
Lung2−5.31−6.99, −3.63<0.0010.00.688F
Kidney1−4.03−6.12, −1.93<0.001--R
 Animal modelMice3−12.17−20.98, −3.370.00793.6<0.001R
Rats4−5.39−6.88, −3.90<0.00119.10.295F
GSH
 Nanomaterials typeZnONPs28.543.04, 14.040.00283.90.013R
TiO2NPs16.964.52, 9.39<0.001--R
GNPs13075.001727.35, 4422.65<0.001--R
GO21.46−0.23, 3.160.09072.30.057R
 Dosage≤100 mg/kg55.162.10, 8.220.00192.7<0.001R
>100 mg/kg13075.001727.35, 4422.65<0.001--R
 Duration≤2 weeks21463.89−1.5 × 103, 4465.950.33995.0<0.001R
>2 weeks44.691.38, 8.010.00592.9<0.001R
 Sample sourceLiver37.744.97, 10.52<0.00167.90.044R
Lung21.46−0.23, 3.160.09072.30.057R
Kidney13075.001727.35, 4422.65<0.001--R
 Animal modelMice33.140.18, 6.100.03790.9<0.001R
Rats35.268.93, −2.180.11592.3<0.001R
TNF-α
 Nanomaterials typeZnONPs5−2.68−6.96, 1.600.21994.7<0.001R
TiO2NPs2−4.53−8.13, −0.930.01484.60.011R
 Dosage≤100 mg/kg6−5.14−6.39, −3.90<0.00157.50.038R
>100 mg/kg110.006.62, 13.38<0.001--R
 Duration≤2 weeks2−4.53−8.13, −0.930.01484.60.011R
>2 weeks5−2.68−6.96, 1.600.21994.7<0.001R
 Sample sourceLiver2−4.53−8.13, −0.930.01484.60.011R
Serum5−2.68−6.96, 1.600.21994.7<0.001R
 Animal modelMice1−6.45−8.74, −4.17<0.001--R
Rats6−2.75−6.10, 0.600.10893.6<0.001R
IL-6
 Nanomaterials typeZnONPs5−13.23−14.18, −20.08<0.00188.9<0.001R
TiO2NPs1−9.88−13.22, −6.53<0.001--R
 Dosage≤100 mg/kg5−14.71−18.50, −10.91<0.00167.00.017R
>100 mg/kg1−5.99−8.13, −3.85<0.001--R
 Duration≤2 weeks1−9.88−13.22, −6.53<0.001--R
>2 weeks5−13.23−14.18, −20.08<0.00188.9<0.001R
 Sample sourceLiver5−13.23−14.18, −20.08<0.00188.9<0.001R
Serum1−9.88−13.22, −6.53<0.001--R
 Animal modelMice 1−9.88−13.22, −6.53<0.001--R
Rats 5−13.23−14.18, −20.08<0.00188.9<0.001R
Caspase-3
 Nanomaterials typeZnONPs4−10.17−10.45, −4.90<0.00191.0<0.001
TiO2NPs1−4.39−6.07, −2.71<0.001--
AgNPs1−0.46−2.09, 1.180.584--
 Duration≤2 weeks1−4.39−6.07, −2.71<0.001--
>2 weeks5−8.01−12.55, −3.470.00193.8<0.001
 Sample sourceLiver3−11.01−18.55, −3.480.00492.6<0.001
Brain1−0.46−2.09, 1.180.584--
Heart2−6.32−11.21, −1.430.01187.60.004
 Animal modelMice1−4.39−6.07, −2.71<0.001--
Rats5−8.01−12.55, −3.470.00193.8<0.001
Tail length
 Nanomaterials typeZnONPs4−5.21−8.60, −1.820.00394.7<0.001R
TiO2NPs1−48.07−63.79, −32.34<0.001--R
 Sample sourceLiver3−3.35−7.19, 0.500.08894.6<0.001R
Heart2−12.05−21.40, −2.700.01288.80.003R
 Animal modelMice 1−48.07−63.79, −32.34<0.001--R
Rats4−5.21−8.60, −1.820.00394.7<0.001R
Tail DNA %
 Nanomaterials typeZnONPs4−0.73−1.42, −0.050.03553.4<0.092R
TiO2NPs1−166.00−220.23, −111.77<0.001--R
 Sample sourceLiver3−1.11−4.79, 2.580.55794.4<0.001R
Heart2−1.19−2.57, 0.180.08974.10.050R
 Animal modelMice1−166.00−220.23, −111.77<0.001--R
Rats4−0.73−1.42, −0.050.03553.4<0.092R
ROS, reactive oxygen species; MDA, malonaldehyde; TNF, tumor necrosis factor; IL, interleukin; TiO2NPs, titanium dioxide nanoparticles; ZnONPs, zinc oxide nanoparticles; CoNPs, cobalt nanoparticles; GNPs, gold nanoparticles; NiONPs, nickel oxide nanoparticles; AgNPs, silver nanoparticles; GO, graphene oxide; SWCNTs, single walled carbon nanotubes; SMD, standardized mean difference; CI, confidence interval; F, fixed-effects; R, random-effects; pH-value, significance for heterogeneity; pE-value, significance for treatment effects. Bold indicated the outcomes significantly changed by vitamins.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xie, D.; Hu, J.; Yang, Z.; Wu, T.; Xu, W.; Meng, Q.; Cao, K.; Luo, X. Vitamin Supplementation Protects against Nanomaterial-Induced Oxidative Stress and Inflammation Damages: A Meta-Analysis of In Vitro and In Vivo Studies. Nutrients 2022, 14, 2214. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14112214

AMA Style

Xie D, Hu J, Yang Z, Wu T, Xu W, Meng Q, Cao K, Luo X. Vitamin Supplementation Protects against Nanomaterial-Induced Oxidative Stress and Inflammation Damages: A Meta-Analysis of In Vitro and In Vivo Studies. Nutrients. 2022; 14(11):2214. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14112214

Chicago/Turabian Style

Xie, Dongli, Jianchen Hu, Zhenhua Yang, Tong Wu, Wei Xu, Qingyang Meng, Kangli Cao, and Xiaogang Luo. 2022. "Vitamin Supplementation Protects against Nanomaterial-Induced Oxidative Stress and Inflammation Damages: A Meta-Analysis of In Vitro and In Vivo Studies" Nutrients 14, no. 11: 2214. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14112214

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