L‑carnitine alleviates oxidative stress‑related damage via MAPK signaling in human lens epithelial cells exposed to H2O2

  • Authors:
    • Xiaoxia Li
    • Fanlan Meng
    • Hua Li
    • Xia Hua
    • Li'an Wu
    • Xiaoyong Yuan
  • View Affiliations

  • Published online on: July 22, 2019     https://doi.org/10.3892/ijmm.2019.4283
  • Pages: 1515-1522
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

L‑carnitine (LC) is well known for its antioxidative properties. The present study aimed to evaluate the effects of LC on human lens epithelial cells (HLECs) and to analyze its regulatory mechanism in cataractogenesis. HLE B‑3 cells were cultured with hydrogen peroxide (H2O2) and were pretreated with or without LC. The Cell Counting kit‑8 assay was used to determine cell viability. Reactive oxygen species (ROS) assay kit was used to measure the cellular ROS production induced by H2O2 and LC. In addition, reverse transcription‑quantitative PCR and western blot analysis were performed to detect the expression levels of oxidative damage markers and antioxidant enzymes. Notably, ROS overproduction was observed upon exposure to H2O2, whereas LC supplementation markedly decreased ROS levels through activation of the antioxidant enzymes forkhead box O1, peroxiredoxin 4 and catalase. Furthermore, LC suppressed the expression of apoptosis‑associated genes (caspase-3) and inflammation‑associated genes [interleukin (IL)1, IL6, IL8 and cyclooxygenase‑2]. Conversely, LC promoted proliferating cell nuclear antigen, cyclin‑dependent kinase (CDK)2 and CDK4 expression, which may increase proliferation of HLECs that were incubated with H2O2. In addition, epithelial‑mesenchymal transition occurred upon ROS accumulation, whereas the effects of H2O2 on AQP1 and vimentin expression were reversed upon LC supplementation. Notably, this study revealed that LC restored the oxidant/antioxidant balance and protected against cell damage through the mitogen‑activated protein kinase signaling pathway. In conclusion, LC may serve a protective role in curbing oxidative damage and therefore may be considered a potential therapeutic agent for the treatment of cataracts.

Introduction

Human lens epithelial cells (HLECs) are vulnerable to oxidative stress, which has deleterious effects on lens transparency and ultimately leads to cataracts. Aberrant reactive oxygen species (ROS) accumulation and scavenging are major contributors to oxidative damage (1-3). Alterations in the cellular microenvironment in response to hydrogen peroxide (H2O2) manifest as apoptosis and result in the production of pro-inflammatory mediators in HLECs (4). The risk of oxidative damage to the transparent lens may be compensated by the presence of antioxidant enzymes. Notably, catalase (CAT) and peroxiredoxins (PRDXs) are antioxidant enzymes that act as ROS scavengers and potential antioxidant protectors against cataract development (5,6). Forkhead box (Fox)O1 belongs to the FoxO family of transcription factors and, when activated, serves a protective role in antioxidative responses (7). Therefore, it is important to establish an antioxidative approach for preventing the formation and progression of cataracts.

Epithelial-mesenchymal transition (EMT) may be initiated by oxidative stress and manifests in the loss of epithelial characteristics and the acquisition of mesenchymal properties (8,9). Aquaporins (AQPs) are intrinsic plasma membrane proteins that possess H2O2 permeability properties. AQP1 protein expression is confined to the lens epithelium, where it acts as an epithelial marker. AQP1 serves a crucial role in the maintenance of ocular lens homeostasis, and its insufficient function may cause cataracts (10). The relationship between EMT and oxidative stress is an important factor in cataract progression.

It is well known that the lens is an organ that lacks nerves (11); therefore, neurodegeneration is not involved in cataract formation. Acetyl-L-carnitine (ALC) is widely used in neurodegenerative diseases due to its neurobiological effects (12). Due to the effects of ALC on neurodegeneration, which is not involved in cataract formation, the present study aimed to explore the effects of the antioxidant L-carnitine (LC) on cataract prevention, not ALC. LC is a water-soluble, vitamin-like molecule that is naturally found in meat; since its recognition, LC has garnered much attention. LC is a pivotal agent involved in protecting the cell and DNA against damage induced by oxidative stress (13,14). LC protects the ocular surface; for example, LC protects against hyperosmotic stress in dry eye disease (15). It has previously been reported that perturbation of the carnitine shuttle by increased plasma levels of long-chain acylcarnitines leads to a compromised cellular capacity to prevent ROS generation in age-related macular degeneration (16). Despite these findings, the connection between LC and cataract prevention remains unclear. The present study aimed to explore the effects of LC on H2O2-induced oxidative damage in HLECs, and to identify the molecular pathways involved in this protection.

Materials and methods

Cell culture

The HLE B-3 cell line was obtained from American Type Culture Collection. The cells were cultured in Dulbecco's modified Eagle's medium/F-12 (HyClone; GE Healthcare Life Sciences) containing 20% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 10 µg/l gentamicin at 37°C in a humidified atmosphere containing 5% CO2. The cells were incubated with 0, 100, 150, 200, 250, 300 and 350 µM H2O2 alone for 24 h, or were pretreated with LC (Sigma-Aldrich; Merck KGaA) at 0, 10, 100, 300, 500 and 700 µM for 16 h. All treatments were carried out at 37°C in a cell culture incubator. In addition, cells were pretreated with LC (500 µM), ERK inhibitor (FR180204, 1.25 µM) or p38 inhibitor (PD169316, 1.25 µM), or a combination of LC and FR180204 or LC and PD169316, for 2 h prior to treatment with 250 µM H2O2 treatment for 24 h. FR180204 and PD169316 were purchased from Sigma-Aldrich; Merck KGaA.

Cell viability assay

The Cell Counting kit-8 assay was used to detect the effects of different concentrations of H2O2 and LC on the viability of HLECs. The optical density (OD) was measured using the CCK-8 method (Dojindo Molecular Technologies, Inc.). Cells were placed in 96-well plates at 2×103 cells/well in 200 µl growth medium and were cultured at 37°C in a humidified incubator with 5% CO2. Following treatment with H2O2 or LC, 10 µl CCK-8 solution was added to each well. After 2 h at 37°C, the absorbance was measured at 450 nm. Cell survival rate was calculated according to the following formula: Cell viability (%)=(As-Ab)/(Ac-Ab), where As is the average OD value of the experimental group, Ab is the average OD value of the blank group, and Ac is the average OD value of the control group.

Measurement of cellular ROS production

Cellular ROS production was detected using a Reactive Oxygen Species Assay kit (cat. no. CA1410; Beijing Solarbio Science & Technology Co., Ltd.). The DCFH-DA ROS probe, which permeates the cell membrane with no fluorescence, was used according to the manufacturer's protocol. ROS induces the production of fluorescent DCF through oxidizing DCFH. Subsequently, ROS levels can be determined by detecting the fluorescence of DCF. Images were captured using a fluorescence microscope (DM4000 B LED; Leica Microsystems GmbH).

Total RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from HLECs using the RNAsimple Total RNA Extraction kit [cat. no. dp419; Tiangen Biotech (Beijing) Co., Ltd.] in accordance with the manufacturer's protocol. The extracted RNA was quantified using a NanoDrop 2000 spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.) and stored at −80°C prior to use. First-strand cDNA was synthesized from 2.0 µg total RNA using a M-MLV Reverse Transcriptase kit (Promega Corporation) according to the manufacturer's protocol, and was then stored at -20°C before use. qPCR was performed using the StepOnePlus™ Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), and SYBR Premix Ex Taq™ (Takara Biotechnology Co., Ltd.). The thermocycling conditions were as follows: 95°C for 3 min, followed by 40 cycles at 95°C for 12 sec and 62°C for 40 sec, and a final dissociation stage at 95°C for 15 sec, 65°C for 1 min and 95°C for 15 sec. GAPDH served as an internal control and was used to detect the expression levels of genes in HLECs. Relative gene expression was calculated using the 2−ΔΔCq fold change method (17). The primer sequences used for RT-qPCR are listed in Table I.

Table I

Primer sequences of all genes used in quantitative PCR.

Table I

Primer sequences of all genes used in quantitative PCR.

GeneForward primerReverse primer
H-GAPDH 5′-TGCCCTCAACGACCACTTTG-3′ 5′-CTGGTGGTCCAGGGGTCTTA-3′
H-PRDX4 5′-GAAGGAACAGCTGTGATCGA-3′ 5′-AGAGCATGCTACCACTTCAGT-3′
H-FOXO1 5′-ATGGCTTGGTGTCTTTCTTTTCT-3′ 5′-TGTGGCTGACAAGACTTAACTCAA-3′
H-CAT 5′-TCTGGAGAAGTGCGGAGATT-3′ 5′-TCCAATCATCCGTCAAAACA-3′
H-Caspase-3 5′-AGCGAATCAATGGACTCTGGA-3′ 5′-GGTTTGCTGCATCGACATCT-3′
H-PCNA 5′-CACTCCACTCTCTTCAACGGT-3′ 5′-ATCCTCGATCTTGGGAGCCA-3′
H-COX2 5′-ATAACGTGAAGGGCTGTCCC-3′ 5′-ATCATCTAGTCCGGAGCGGG-3′
H-IL1 5′-TCATACCAAGGAGAAGTAATAAGCC-3′ 5′-ACCAAAGAAGTACAGCGCCAT-3′
H-IL6 5′-CAAACTTCCTGGAGTTCACC-3′ 5′-TGTGTCCAATGGACAGGATG-3′
H-IL8 5′-ACCTCACTGTGCAAATTCAG-3′ 5′-TATGACTCTTGCTGCTCAGC-3′
H-AQP1 5′-TAACCCTGCTCGGTCCTTTG-3′ 5′-AGTCGTAGATGAGTACAGCCAG-3′
H-CDK2 5′-AAATTCATGGATGCCTCTGC-3′ 5′-CAGGGACTCCAAAAGCTCTG-3′
H-CDK4 5′-GAACTGACCGGGAGATCAAG-3′ 5′-TCAGATCTCGGTGAACGATG-3′
H-α-SMA 5′-TGACATTTGTGAAACTTCGGGT-3′ 5′-TGAAGCAATGGTAGCTGGGT-3′
H-Vimentin 5′-AAATGGCTCGTCACCTTCGT-3′ 5′-AGAAATCCTGCTCTCCTCGC-3′

[i] α-SMA, α-smooth muscle actin; AQP1, aquaporin 1; CAT, catalase; CDK, cyclin-dependent kinase; COX2, cyclooxygenase-2; H, human; IL, interleukin; PCNA, proliferating cell nuclear antigen; PRDX4, peroxiredoxin 4.

Western blot analysis

Cells treated with H2O2 and LC were homogenized using RIPA lysis buffer with protease and phosphatase inhibitor cocktail (Beyotime Institute of Biotechnology). Protein samples were quantified using the bicinchoninic acid protein assay (Thermo Fisher Scientific, Inc.). Equal quantities of proteins per lane (30-50 µg, according to the thickness of the gel) were separated by 10-12% SDS-PAGE, transferred onto PVDF membranes and were then incubated with 5% blocking reagent (5% skim milk) for 1.5 h at room temperature. Subsequently, the membranes were incubated with the following antibodies overnight at 4°C: Anti-GAPDH (1:1,000; cat. no. ab181602), anti-PRDX4 (1:1,000; cat. no. ab184167), anti-cleaved caspase-3 (1:1,000; cat. no. ab2302), anti-interleukin (IL)-1β (1:1,000; cat. no. ab45692), anti-vimentin (1:1,000; cat. no. ab92547) and anti-cyclooxygenase-2 (COX2; 1:1,000; cat. no. ab179800) (all from Abcam); anti-proliferating cell nuclear antigen (PCNA; 1:1,000; cat. no. 13110), anti-ERK (1:1,000; cat. no. 4695), anti-phosphorylated (P)-ERK (1:1,000; cat. no. 4370), anti-p38 (1:1,000; cat. no. 8690) and anti-p-p38 (1:1,000; cat. no. 4511) (all from Cell Signaling Technology, Inc.). The membranes were washed three times with phosphate buffered saline-0.1% Tween-20 (Beijing Solarbio Science & Technology Co., Ltd.), and were then incubated with horseradish peroxidase-conjugated secondary antibodies (1:10,000; cat. no. ab6721; Abcam) for 2 h at room temperature. Blots were detected using enhanced chemiluminescence reagents (EMD Millipore) and were exposed to chemiluminescent film (Kodak) or using G:BOX F3 (Syngene). The images were analyzed using ImageJ software (v.1.42q; National Institutes of Health).

Statistical analysis

Data are presented as the mean ± standard error of the mean from three independent experiments. The significance of differences between two groups was evaluated using two-tailed Student's t-test, and one-way ANOVA (significant variation was indicated as α=0.05) followed by Dunnett's multiple comparisons test or Tukey's multiple comparisons test were used to evaluate the significance of differences among multiple groups. GraphPad Prism version 7.0 software (GraphPad Software, Inc.) was used to conduct statistical analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

LC reverses the effects of H2O2 on viability of HLE B-3 cells

H2O2 induced a significant decrease in HLE B-3 cell viability in a dose-dependent manner (Fig. 1A). Subsequently, 250 µM was chosen as the optimal concentration in the subsequent experiments, because it was approximately equal to the IC50 of H2O2. Conversely, treatment with LC induced minor alterations in cell viability (Fig. 1B), indicating the minimal cytotoxicity of LC. In addition, LC exerted an ameliorating effect on H2O2-induced suppression of cell viability; however, this effect was not dose-dependent (Fig. 1C). Notably, cell viability was reduced to some extent when exposed to 700 µM LC alone; therefore, LC concentrations at 100, 300 and 500 µM were chosen for subsequent experiments.

LC decreases the generation of ROS in HLECs and promotes antioxidant production

To determine the role of LC in ROS-induced oxidative damage, HLE B-3 cells were exposed to H2O2 with or without LC pretreatment. This study aimed to determine whether exposure to H2O2 and LC could modify ROS generation. A marked increase in DCF-positive cells was observed by fluorescence microscopy in HLE B-3 cells exposed to H2O2, as shown in Fig. 2A. DCF fluorescence was markedly reduced by LC pretreatment, thus suggesting that LC partially restrained H2O2-induced ROS generation in cells induced by H2O2.

As shown in Fig. 2B, compared with in the control group, the mRNA expression levels of FoxO1 exhibited a ~50% reduction in response to H2O2 treatment, but were increased following treatment with LC. Similarly, compared with in the H2O2 group, the mRNA expression levels of the antioxidant enzymes PRDX4 and CAT were increased upon exposure of HLE B-3 cells to LC (P<0.05). Similar results were obtained by western blotting to detect PRDX4 protein expression (Fig. 2C). These findings indicated that LC may exert protective effects on cells suffering from oxidative damage.

LC inhibits H2O2-induced increase of apoptosis-associated and inflammation-associated genes

Cleaved-caspase-3 was detected as a marker of apoptosis; its expression was increased in HLECs exposed to H2O2. Conversely, pretreatment with LC partially reversed the increase in cleaved-caspase-3 mRNA and protein expression (Fig. 3A and B; P<0.05). Notably, compared with in the control group, H2O2 exposure induced a ~1.6-fold increase in cleaved-caspase-3 protein expression levels, as determined by western blotting (Fig. 3B).

The mRNA expression levels of inflammatory markers COX2, IL1, IL6 and IL8 were increased with H2O2 exposure (Fig. 3A), indicating the possible involvement of inflammation during cataract progression. LC reversed the inflammatory reaction induced by H2O2 exposure; however, the effects were not dose-dependent. Western blot analysis revealed that the protein expression levels of IL-1β were increased following H2O2 treatment, whereas these levels were reduced by LC pretreatment (Fig. 3B). Taken together, these data indicated that LC may have a role in reducing H2O2-induced apoptosis via alleviating inflammatory responses.

LC restores proliferation and suppresses ROS-induced EMT in HLECs

The expression levels of EMT-associated genes were detected in HLE B-3 cells exposed to H2O2. The expression levels of AQP1, an epithelial marker, were reduced by H2O2. Conversely, the expression levels of the mesenchymal markers vimentin and α-smooth muscle actin (α-SMA) were increased in the H2O2 group (P<0.05). Conversely, LC pretreatment significantly reversed the expression patterns of the aforementioned genes at the mRNA level (P<0.05; Fig. 4A). Western blot analysis further verified the effects of H2O2 and LC on the protein expression levels of vimentin, thus indicating that LC inhibited ROS-induced EMT (Fig. 4B).

Subsequently, the modulatory effects of LC on proliferative markers were analyzed. PCNA expression was decreased to ~70% of the level in the control group in response to H2O2 exposure (P<0.05). LC pretreatment increased PCNA expression at the mRNA and protein levels compared with in the H2O2 group (Fig. 5A and B). CDK2 and CDK4 mRNA expression was reduced upon H2O2 exposure, whereas LC restored their expression (Fig. 5A; P<0.05).

LC regulates oxidative damage through the mitogen-activated protein kinase (MAPK) pathway

Intracellular ROS activates p38 MAPK, an oxidative sensor that belongs to the MAPK family (18); therefore, to decipher the potential cellular mechanism underlying the effects of LC on oxidative damage, this study evaluated whether the MAPK signaling pathway was involved. As determined by western blot analysis (Fig. 5C), p-ERK1, p-ERK2 and p-p38 were significantly enhanced by H2O2; however, they were significantly reduced by LC in a dose-dependent manner (P<0.05), thus suggesting the involvement of the MAPK pathway in LC-modified oxidative stress.

To further evaluate the role of the MAPK signaling pathway in mediating the protective effects of LC on H2O2-induced cell inhibition and inflammation, cell viability and COX-2 expression was assessed in cells exposed to H2O2 and LC in the presence of an ERK inhibitor (FR180204) or p38 inhibitor (PD169316). As shown in Fig. 5D, H2O2 markedly enhanced COX-2 expression, whereas LC reversed COX-2 expression. Furthermore, pretreatment with FR180204 or PD169316 abolished H2O2-induced COX-2 expression. In addition, HLE B-3 cells exposed to H2O2 and LC combined with FR180204 or PD169316 exhibited considerably increased cell viability (Fig. 5E). These results indicated that LC may exert beneficial effects against oxidative damage via MAPK signaling.

Discussion

Oxidative stress is a risk factor for cataracts caused by the overproduction of ROS. H2O2 is a main type of ROS that leads to oxidative damage in HLECs. Antioxidants that scavenge excess ROS serve as a defense against cell damage (19). In this study, it was demonstrated that LC exhibited minimal cytotoxicity and reversed H2O2-induced ROS production.

Exposure of HLE B-3 cells to H2O2 triggered oxidative damage, which was reflected in the destructed antioxidant defense mechanism. Antioxidant substances, including FoxO1, PRDX4 and CAT, are involved in ROS scavenging and serve as potential protectors. H2O2-induced oxidative damage is associated with decreased FoxO1, PRDX4 and CAT activities (20,21), indicating the probable mechanisms underlying cataract formation. The present study focused on the oxidative damage caused by ROS imbalance; therefore, FoxO1, PRDX4 and CAT were detected as antioxidative substances.

FoxO1 is highly expressed as a downstream antioxidant when activated (22). The present study revealed that FoxO1 is highly expressed in HLECs treated with LC, suggesting that LC possesses antioxidative potential. The present results indicated that LC may exert beneficial effects on ROS scavenging by increasing the expression levels of the antioxidative enzymes CAT and PRDX4. This finding is consistent with previous findings, which suggested that LC may protect retinal pigment epithelial cells from H2O2-induced oxidative damage by increasing antioxidant and antioxidant enzyme activity (23). This study hypothesized that LC may act as a potential antioxidant protector against cataract formation. Our future study aims to further explore alterations in transcriptional regulation and the potential underlying mechanism. In addition, further studies are required to determine the optimal therapeutic delivery method of LC to the lens. Notably, widely used topical inserts and colloidal drug delivery systems (24), such as nanowafers (25), may represent possible pharmacological vehicles to enhance therapeutic efficacy.

PCNA is an auxiliary protein that facilitates cell cycle progression. CDK2 and CDK4 are well-known stimulators and promoters of the cell cycle from the G0/G1 into S phase (26,27). In this study, PCNA, CDK2 and CDK4 expression were enhanced by LC in the presence of H2O2, demonstrating the role of LC in protecting HLECs against oxidative damage. Given its antioxidant properties, LC may promote the cell cycle and thereby increase cell proliferation.

Exposure to H2O2 may promote EMT in the transparent lens. In this study, marked decreases in the expression of the epithelial marker AQP1, together with an increase in mesenchymal markers (vimentin and α-SMA), were observed in HLECs exposed to H2O2. Prevention of EMT was demonstrated by elevated AQP1 expression, and attenuated vimentin and α-SMA expression in the presence of LC. This result is consistent with a previous study suggesting that LC prevents the expression of EMT-associated biomarkers in renal fibrosis (28). It was hypothesized that LC may become activated in response to ROS production and scavenging; however, the exact mechanisms require further analysis.

Oxidative stress is closely associated with inflammatory processes, which are important for the initiation and progression of cataracts (29,30). The expression of proinflammatory cytokines, including IL-1β, IL6 and IL8, was reduced by LC pretreatment. COX2 is a major oxygenase, and its expression increases along with oxidative stress-induced inflammation (31). This study further revealed that the production of COX2 was markedly induced by H2O2, but significantly rescued by LC. Inflammation triggers LECs to undergo an apoptotic response and subsequently initiate cataract formation (32). Cleaved-caspase-3 expression was decreased and inflammation was inhibited upon LC exposure. In the present study, it was revealed that H2O2 may act as a mediator of inflammation and apoptosis in HLECs, whereas LC could significantly attenuate inflammation and reduce apoptosis of HLECs.

The well-known MAPK signaling pathway, which includes p38/MAPK, ERK and JNK, is involved in regulating oxidative damage when cataracts occurs (33,34). The present results indicated that ERK/MAPK and p38/MAPK were significantly activated upon H2O2 exposure, whereas LC significantly reduced the phosphorylation of ERK and p38 induced by H2O2. Therefore, it was hypothesized that the p38/MAPK and ERK/MAPK pathways are involved in the protective mechanism underlying the effects of LC on oxidative damage in HLECs. It has been reported that MAPK pathway inhibitors can regulate apoptosis and inflammatory responses. The present results revealed that ERK and p38 inhibitors significantly reduced H2O2-induced cytotoxicity and inhibited the expression of the inflammatory cytokine COX2 induced by exposure to H2O2. These findings provide insight into how oxidative modification of LC contributes to cataract prevention.

In conclusion, the protective effects of LC against oxidative stress may be attributed to its ROS-scavenging ability. Oxidative damage in HLECs may be reversed by LC, which prevents the induction of inflammation, apoptosis and EMT through the p38/MAPK and ERK/MAPK pathways. The obtained results suggested that LC may serve an important role in protecting HLECs from peroxidative damage and may be a promising therapeutic modality for the treatment of cataracts.

Acknowledgments

The authors would like to thank Ms. Ruifang Han, Mr. Ming Ying and Mr. Peng Hao (Tianjin Key Laboratory of Ophthalmology and Visual Science) for their technical assistance.

Funding

This study was supported in part by the National Nature Science Foundation of China (grant no. 81670817); the Tianjin Research Program of Application Foundation and Advanced Technology (grant no. 17JCYBJC27200); the Science & Technology Foundation for Selected Overseas Chinese Scholar, Bureau of Personnel of China, Tianjin; Talent Innovation Group of 131, Bureau of Personnel, Tianjin; Tianjin Science and Technology Project (Popularization of Science grant no. 17KPHDSF00230), and China Postdoctoral Science Foundation (grant no. 2018M641665).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XY and HL made substantial contributions to the concept and design of the present study. XL, FM, XH and LW performed the experiments. XL analyzed the data and wrote the paper. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Nagai N, Ito Y and Takeuchi N: Correlation between hyper-sensitivity to hydrogen peroxide and low defense against Ca(2+) influx in cataractogenic lens of Ihara cataract rats. Biol Pharm Bull. 34:1005–1010. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Zuercher J, Neidhardt J, Magyar I, Labs S, Moore AT, Tanner FC, Waseem N, Schorderet DF, Munier FL, Bhattacharya S, et al: Alterations of the 5′ untranslated region of SLC16A12 lead to age-related cataract. Invest Ophthalmol Vis Sci. 51:3354–3361. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Chang D, Zhang X, Rong S, Sha Q, Liu P, Han T and Pan H: Serum antioxidative enzymes levels and oxidative stress products in age-related cataract patients. Oxid Med Cell Longev. 2013:5878262013. View Article : Google Scholar : PubMed/NCBI

4 

Mok JW, Chang DJ and Joo CK: Antiapoptotic effects of anthocyanin from the seed coat of black soybean against oxidative damage of human lens epithelial cell induced by H2O2. Curr Eye Res. 39:1090–1098. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Fujii J, Ikeda Y, Kurahashi T and Homma T: Physiological and pathological views of peroxiredoxin 4. Free Radic Biol Med. 83:373–379. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Yamada S and Guo X: Peroxiredoxin 4 (PRDX4): Its critical in vivo roles in animal models of metabolic syndrome ranging from atherosclerosis to nonalcoholic fatty liver disease. Pathol Int. 68:91–101. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Kim DH, Park CH, Park D, Choi YJ, Park MH, Chung KW, Kim SR, Lee JS and Chung HY: Ginsenoside Rc modulates Akt/FoxO1 pathways and suppresses oxidative stress. Arch Pharm Res. 37:813–820. 2014. View Article : Google Scholar

8 

Kubo E, Shibata T, Singh DP and Sasaki H: Roles of TGF β and FGF signals in the lens: Tropomyosin regulation for posterior capsule opacity. Int J Mol Sci. 19:E30932018. View Article : Google Scholar

9 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Schey KL, Petrova RS, Gletten RB and Donaldson PJ: The role of aquaporins in ocular lens homeostasis. Int J Mol Sci. 18:E26932017. View Article : Google Scholar : PubMed/NCBI

11 

Dahm R: Dying to see. Sci Am. 291:82–89. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Traina G: The neurobiology of acetyl-L-carnitine. Front Biosci (Landmark Ed). 21:1314–1329. 2016. View Article : Google Scholar

13 

Nutrients Editorial Office: Erratum: l-Carnitine supplementation in recovery after exercise; Nutrients 2018, 10, 349. Nutrients. 10:E5412018. View Article : Google Scholar

14 

Mishra A, Reddy IJ, Gupta PS and Mondal S: L-carnitine mediated reduction in oxidative stress and alteration in transcript level of antioxidant enzymes in sheep embryos produced in vitro. Reprod Domest Anim. 51:311–321. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Deng R, Su Z, Hua X, Zhang Z, Li DQ and Pflugfelder SC: Osmoprotectants suppress the production and activity of matrix metalloproteinases induced by hyperosmolarity in primary human corneal epithelial cells. Mol Vis. 20:1243–1252. 2014.PubMed/NCBI

16 

Mitchell SL, Uppal K, Williamson SM, Liu K, Burgess LG, Tran V, Umfress AC, Jarrell KL, Cooke Bailey JN, Agarwal A, et al: The carnitine shuttle pathway is altered in patients with neovascular age-related macular degeneration. Invest Ophthalmol Vis Sci. 59:4978–4985. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

18 

Werner E, Wang H and Doetsch PW: Opposite roles for p38MAPK-driven responses and reactive oxygen species in the persistence and resolution of radiation-induced genomic instability. PLoS One. 9:e1082342014. View Article : Google Scholar : PubMed/NCBI

19 

Bai J, Yang F, Dong L and Zheng Y: Ghrelin protects human lens epithelial cells against oxidative stress-induced damage. Oxid Med Cell Longev. 2017:19104502017. View Article : Google Scholar : PubMed/NCBI

20 

Akasaki Y, Alvarez-Garcia O, Saito M, Caramés B, Iwamoto Y and Lotz MK: FoxO transcription factors support oxidative stress resistance in human chondrocytes. Arthritis Rheumatol. 66:3349–3358. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Patel H, Chen J and Kavdia M: Induced peroxidase and cytoprotective enzyme expressions support adaptation of HUVECs to sustain subsequent H2O2 exposure. Microvasc Res. 103:1–10. 2016. View Article : Google Scholar

22 

Zhu L, Li J, Wu D and Li B: The protective effect of beta-casomor-phin-7 via promoting Foxo1 activity and nuclear translocation in human lens epithelial cells. Cutan Ocul Toxicol. 37:267–274. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Shamsi FA, Chaudhry IA, Boulton ME and Al-Rajhi AA: L-carnitine protects human retinal pigment epithelial cells from oxidative damage. Curr Eye Res. 32:575–584. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Thrimawithana TR, Rupenthal ID, Räsch SS, Lim JC, Morton JD and Bunt CR: Drug delivery to the lens for the management of cataracts. Adv Drug Deliv Rev. 126:185–194. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Yuan X, Marcano DC, Shin CS, Hua X, Isenhart LC, Pflugfelder SC and Acharya G: Ocular drug delivery nanowafer with enhanced therapeutic efficacy. ACS Nano. 9:1749–1758. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Zhao C, Fu MJ and Qiu LH: Molecular cloning and functional characterization of cyclin E and CDK2 from penaeus monodon. Genet Mol Res. 15:2016. View Article : Google Scholar

27 

Kanska J, Zakhour M, Taylor-Harding B, Karlan BY and Wiedemeyer WR: Cyclin E as a potential therapeutic target in high grade serous ovarian cancer. Gynecol Oncol. 143:152–158. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Chou HC, Wen LL, Chang CC, Lin CY, Jin L and Juan SH: From the cover: l-Carnitine via PPARγ- and Sirt1-dependent mechanisms attenuates epithelial-mesenchymal transition and renal fibrosis caused by perfluorooctanesulfonate. Toxicol Sci. 160:217–229. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Baierle M, Nascimento SN, Moro AM, Brucker N, Freitas F, Gauer B, Durgante J, Bordignon S, Zibetti M, Trentini CM, et al: Relationship between inflammation and oxidative stress and cognitive decline in the institutionalized elderly. Oxid Med Cell Longev. 2015:8041982015. View Article : Google Scholar : PubMed/NCBI

30 

Dogru M, Kojima T, Simsek C and Tsubota K: Potential role of oxidative stress in ocular surface inflammation and dry eye disease. Invest Ophthalmol Vis Sci. 59:DES163–DES168. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Hua X, Chi W, Su L, Li J, Zhang Z and Yuan X: ROS-induced oxidative injury involved in pathogenesis of fungal keratitis via p38 MAPK activation. Sci Rep. 7:104212017. View Article : Google Scholar : PubMed/NCBI

32 

Xu D, Zhu H, Fu Q, Xu S, Sun W, Chen G and Lv X: Ketamine delays progression of oxidative and damaged cataract through regulating HMGB-1/NF-κB in lens epithelial cells. Immunopharmacol Immunotoxicol. 40:303–308. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Jia Z, Song Z, Zhao Y, Wang X and Liu P: Grape seed proanthocyanidin extract protects human lens epithelial cells from oxidative stress via reducing NF-κB and MAPK protein expression. Mol Vis. 17:210–217. 2011.PubMed/NCBI

34 

Yao K, Ye P, Zhang L, Tan J, Tang X and Zhang Y: Epigallocatechin gallate protects against oxidative stress-induced mitochondria-dependent apoptosis in human lens epithelial cells. Mol Vis. 14:217–223. 2008.PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 44 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li X, Meng F, Li H, Hua X, Wu L and Yuan X: L‑carnitine alleviates oxidative stress‑related damage via MAPK signaling in human lens epithelial cells exposed to H2O2. Int J Mol Med 44: 1515-1522, 2019
APA
Li, X., Meng, F., Li, H., Hua, X., Wu, L., & Yuan, X. (2019). L‑carnitine alleviates oxidative stress‑related damage via MAPK signaling in human lens epithelial cells exposed to H2O2. International Journal of Molecular Medicine, 44, 1515-1522. https://doi.org/10.3892/ijmm.2019.4283
MLA
Li, X., Meng, F., Li, H., Hua, X., Wu, L., Yuan, X."L‑carnitine alleviates oxidative stress‑related damage via MAPK signaling in human lens epithelial cells exposed to H2O2". International Journal of Molecular Medicine 44.4 (2019): 1515-1522.
Chicago
Li, X., Meng, F., Li, H., Hua, X., Wu, L., Yuan, X."L‑carnitine alleviates oxidative stress‑related damage via MAPK signaling in human lens epithelial cells exposed to H2O2". International Journal of Molecular Medicine 44, no. 4 (2019): 1515-1522. https://doi.org/10.3892/ijmm.2019.4283