Protective effects of rosmarinic acid against hydrogen peroxide‑induced cellular senescence and the inflammatory response in normal human dermal fibroblasts

  • Authors:
    • Hyung Jin Hahn
    • Ki Bbeum Kim
    • In‑Sook An
    • Kyu Joong Ahn
    • Hyun Joo Han
  • View Affiliations

  • Published online on: October 17, 2017     https://doi.org/10.3892/mmr.2017.7804
  • Pages: 9763-9769
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hydrogen peroxide (H2O2) is a reactive oxygen species (ROS) that induces numerous cellular events, including cellular senescence and inflammatory responses. Therefore, the aim of this study was to investigate the protective effect of Rosmarinic acid (RA) in H2O2‑induced oxidative stress in normal human dermal fibroblasts (NHDFs). Cytotoxicity assays were performed using a water‑soluble tetrazolium salt, and senescence‑associated β‑galactosidase activity was determined to investigate the proportion of senescent cells. Antioxidant capacities were evaluated via H2O2‑scavenging activity, reverse transcription‑quantitative polymerase chain reaction, NRF2 luciferase reporter gene activity and intracellular ROS scavenging assays. Cytokine‑coded gene expression analysis and nuclear factor‑κB luciferase activity were determined to verify the anti‑inflammatory effect of RA. As a result, the present study demonstrated that rosmarinic acid inhibited H2O2‑induced oxidative stress and inflammatory responses in normal human dermal fibroblasts. Initially, the doses of RA that exerted minimal cytotoxic effects in NHDFs were determined using a cytotoxicity assay. Subsequently, pretreatment with the appropriate doses of RA significantly reversed the H2O2‑induced decrease in NHDF cell viability and decreased cellular senescence of NHDFs. In addition, RA inhibited H2O2‑induced ROS production in NHDFs, as determined by a ROS scavenging assay. The protective effects of RA were mediated by the inhibition of nuclear factor erythroid‑derived 2‑like 2, a transcription factor that functions as a key regulator of redox sensitivity. Furthermore, RA suppressed H2O2‑induced inflammation in NHDFs and significantly rescued H2O2‑induced downregulation of sirtuin 1. RA also inhibited nuclear factor (NF)‑κB transcriptional activity and the expression of NF‑κB target genes, including tumor necrosis factor‑α and interleukin‑6, in H2O2‑exposed NHDFs. Taken together, these data indicate that RA inhibits H2O2‑induced cellular damage in NHDFs.

Introduction

The term reactive oxygen species (ROS) refers to all chemically reactive molecules containing an oxygen atom, including oxygen radicals and non-radicals. The various types of ROS are classified as either endogenous mitochondrial factors or extracellular factors (1). Hydrogen peroxide (H2O2) is a reactive byproduct of the electron transport chain in mitochondria (1).

The skin is the largest organ of the body; it surrounds the body and protects it from the external environment. Extrinsic stimuli, including ultraviolet (UV) light, pollution and thermal stress, disrupt skin cell metabolism, thereby disrupting redox state equilibrium (2). Although cells employ efficient enzymatic and nonenzymatic antioxidant mechanisms, excessive ROS production can induce lipid peroxidation, DNA damage and protein denaturation (1). Proteins associated with conventional enzyme-mediated antioxidant mechanisms include superoxide dismutase (SOD), glutathione peroxidase and catalase (CAT) (1). These enzymes are primarily activated in response to ROS. In H2O2-exposed normal human dermal fibroblasts (NHDFs), excessive ROS production disrupts antioxidant defense mechanisms and induces the inflammatory response (1,3).

In its inactive form in the cytoplasm, nuclear factor erythroid-derived 2-like 2 (NRF2) interacts with Kelch-like-ECH-associated protein 1 (4). A disruption in this interaction by factors, including ROS, activates the NRF2 signaling pathway (4). Activated NRF2 translocates to the nucleus where it binds to antioxidant response element (ARE), and initiates the transcription of genes associated with the response to oxidative stress, including heme oxygenase-1 (HO-1), SOD and CAT (57).

The mammalian Sir2 ortholog, sirtuin 1 (SIRT1), is a histone deacetylase that targets histones and nonhistone substrates, including nuclear factor (NF)-κB and p53 (8,9). Previous studies have demonstrated that SIRT1 is associated with the regulation of numerous cellular processes, including inflammation, apoptosis and autophagy (1012). Under conditions of adaptive stress, including hypoxia, infection and caloric restriction, SIRT1 inhibits the proinflammatory cytokine NF-κB (13). NF-κB serves a dominant role in inducing the inflammatory response (14,15). In response to excessive cellular stress, activated NF-κB directly activates the expression of genes encoding key inflammatory cytokines, including interleukin (IL)-6 and tumor necrosis factor (TNF)-α (16).

The use of the plant secondary metabolite rosmarinic acid (RA) has been studied in pharmaceutical and dietary supplements in Alzheimer's disease, atopic dermatitis and cardiovascular disease (1721). Previous research has revealed that the effects of RA in these contexts are mediated by its antioxidant properties. One report demonstrated that the antioxidant activity of RA is able to inhibit UV-induced damage in the skin of mice (22). In addition, RA may protect human melanoma cells from H2O2-induced oxidative stress (23). Other reports have also demonstrated that RA protects human keratinocyte HaCaT cells from UVB-induced oxidative stress (24,25). However, the effects of RA on NHDFs, which are the key mediators of skin firmness and elasticity, remain unclear. The present study aimed to investigate the effects of RA on H2O2-exposed NHDFs.

Materials and methods

Cell culture and treatment

NHDFs (Lonza Group, Ltd., Basel, Switzerland) and the NF-κB Luciferase Reporter NIH3T3 Stable Cell Line (Signosis, Inc., Santa Clara, CA, USA) were cultured in Dulbecco's modified Eagle's medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 1% penicillin/streptomycin (Thermo Fisher Scientific, Inc.) at 37°C in an atmosphere containing 5% CO2. H2O2 (Sigma-Aldrich; Merck KGaA) was diluted in phosphate-buffered saline (PBS) to obtain a 1 M stock solution. For the H2O2 treatment experiments, the H2O2 stock solution was diluted in cell culture media at the indicated concentrations. RA (Sigma-Aldrich; Merck KGaA) was dissolved in dimethyl sulfoxide (Sigma-Aldrich; Merck KGaA).

Cell viability assay

Cytotoxicity was evaluated using a water-soluble tetrazolium salt (WST-1) assay (EZ-Cytox Cell Viability Assay kit; Itsbio, Seoul, Korea). NHDFs were seeded on 96-well culture plates at a density of 3×103 cells and were incubated for 24 h, at 37°C. Subsequently, the cells were incubated with RA (0–50 µM) for 12 h, after which the cytotoxicity of RA was assessed. To examine the effects of RA against H2O2-induced cytotoxicity, NHDFs were incubated with the indicated concentration of RA (10, 20 and 30 µM) for 12 h prior to H2O2 treatment. After 12 h, 600 µM of H2O2 was added and incubated at 37°C for 2 h. 1/10 volume of WST-1 solution was then added to each well, and the cells were incubated at 37°C for 1 h. Cell viability was evaluated by measuring absorbance at 450 nm using an iMark microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

H2O2 scavenging activity assay

The H2O2 scavenging activity of RA was evaluated as previously described (26). A solution containing H2O2 (40 mM in distilled water, pH 7.4) and varying concentrations of RA (0–1 mg/ml) was incubated at room temperature for 10 min. Scavenger activity was subsequently assessed by measuring the absorbance of the H2O2/RA solution at 230 nm using a UV spectrophotometer (Shimadzu Corporation, Kyoto, Japan). A solution lacking H2O2 was used as a control, and L-ascorbic acid was used as the experimental control treatment. The experiments were conducted in triplicate at each concentration of RA. Scavenger activity was calculated using the following formula: H2O2 scavenging activity (%) = (1 - sample/control) × 100.

Intracellular ROS scavenging assay

Intracellular ROS production was evaluated using a 2′7′-dichlorofluorescein diacetate (DCF-DA; Sigma-Aldrich; Merck KGaA) staining assay. Briefly, cells were incubated with DCF-DA solution (100 µM) at 37°C for 1 h, after RA and H2O2 treatment. ROS levels were then analyzed using flow cytometry. The proportion of fluorescence-positive cells was measured using a FACSCalibur flow cytometer (BD Biosciences, San Diego, CA, USA) with excitation and emission filters of 488 and 530 nm, respectively. N-acetyl-cysteine (NAC), ROS scavenger used as a positive control, was purchased from Sigma-Aldrich; Merck KGaA.

NF-κB luciferase reporter assay

The NF-κB luciferase reporter assay was conducted as previously described (27) with some modifications. Briefly, NF-κB reporter NIH3T3 cells were seeded in 12-well cell culture plates at a density of 2×105 cells/well. After 24 h incubation, cells were pretreated RA for 12 h at 10, 20 and 30 µM, and were then treated with 600 µM H2O2 for 2 h. At the end of the experiments, the cells were resuspended in Passive Lysis Buffer (Promega Corporation, Madison, WI, USA). Subsequently, luciferin (Sigma-Aldrich; Merck KGaA) was added to the cells, and luciferase activity was measured using a Veritas luminometer (Turner Designs, Sunnyvale, CA, USA). The luciferase activity was normalized to β-galactosidase (β-gal) activity, and relative activity is presented as the percentage activity of the control with standard deviation. The results represent the mean of three independent experiments.

Senescence-associated β-gal (SA-β-gal) activity

β-gal expression was used as a marker of senescence (28). β-gal expression levels were determined using an SA-β-gal staining kit (Biovision, Inc., Milpitas, CA, USA) according to the manufacturer's instructions. NHDFs were seeded at a density of 1×106 cells/well in 60-mm cell culture plates and were incubated until cell confluence reached 90%. Later, cells were pretreated RA for 12 h at 10, 20 and 30 µM, and were then treated with 600 µM H2O2 for 2 h. After treatment, the cells were washed in PBS and incubated in 0.5 ml fixative solution (4% formaldehyde, 0.5% glutaraldehyde in PBS buffer, pH 7.2) for 10 min at room temperature. The fixed cells were incubated in a staining solution mixture [staining solution (470 µl), staining supplement (5 µl) and 20 mg/ml X-Gal in dimethylformamide (25 µl)] for 24 h at 37°C. Subsequently, 70% glycerol (1 ml) was added to the cells and blue SA-β-gal-positive cells were counted under a microscope (Olympus IX51; Olympus Corporation, Tokyo, Japan).

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Before RNA extraction, cells were treated with RA (10, 20 and 30 µM) for 12 h and H2O2 (600 µM) for 2 h, in a consecutive manner. Total RNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. RNA purity and concentration were evaluated using a MaestroNano® microspectrophotometer (Maestrogen, Las Vegas, NV, USA). Total RNA was reverse transcribed into cDNA using the oiligo d(T)23 primer and ProtoScript First Strand cDNA Synthesis kit (New England BioLabs, Inc., Ipswich, MA, USA) following the manufacturer's instructions. HO-1, SOD1, CAT, SIRT1, TNF-α and IL-6 mRNA expression levels were evaluated using 5× HOT FIREPOL® EvaGreen® qPCR mix (Solis BioDyne, Tartu, Estonia), a StepOnePlus Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) and gene-specific primers. Each thermo-cycling condition was same in denaturation (95°C) and extension (72°C), except the annealing temperature which differed when different primers were used. The Cq value associated with each gene was normalized to the expression of the β-actin housekeeping gene. The sequences of the qPCR primers are provided in Table I. The 2−ΔΔCq method (29) was used to calculate the relative expression levels of each gene.

Table I.

Primer sequences for quantitative polymerase chain reaction.

Table I.

Primer sequences for quantitative polymerase chain reaction.

Gene nameGene symbolForward (5′-3′)Reverse (5′-3′)
Superoxide dismutase 1SOD1 GGGAGATGGCCCAACTACTG CCAGTTGACATGCAACCGTT
CatalaseCAT ATGGTCCATGCTCTCAAACC CAGGTCATCCAATAGGAAGG
Sirtuin 1SIRT1 GCAGGTTGCGGGAATCCAA GGCAAGATGCTGTTGCAAA
Tumor necrosis factor-αTNF-α CCCAGGGACCTCTCTCTAATC GGTTTGCTACAACATGGGCTACA
Interleukin-6IL-6 TAACAGTTCCTGCATGGGCGGC AGGACAGGCACAAACACGCACC
Actin, betaβ-actin GGATTCCTATGTGGGCGACGA CGCTCGGTGAGGATCTTCATG
Analysis of luciferase reporter gene activity

Transcriptional activity of NRF2 was determined using an antioxidant ARE Reporter kit (cat. no. 60514; BPS Bioscience, San Diego, CA, USA). The luciferase activity was quantitatively assessed according to the manufacturer's instructions. Briefly, NHDFs were cotransfected with NRF2-ARE-luciferase reporter plasmid (BPS Bioscience) and a plasmid that constitutively expressed Renilla luciferase using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). The transfection was conducted according to the manufacturer's instructions with some modifications. The Lipofectamine reagent was mixed with serum-free Opti-MEM (Invitrogen; Thermo Fisher Scientific, Inc.) for 5 min, and then added to the plasmid with gently flicking for 20 min, at room temperature. The transfection mixture was added to the cells and incubated at 37°C. After 24 h of transfection, cells were pretreated for 12 h with the indicated doses of RA (10, 20 and 30 µM), and were then exposed to 600 µM H2O2 for 2 h. The cells were re-suspended in Passive lysis buffer (Promega Corporation) and luciferase activity was measured using a Dual-Luciferase Reporter Assay system (Promega Corporation). The luciferase activity of ARE was normalized to Renilla luciferase activity.

Statistical analysis

All results are presented as the mean ± standard deviation of three independent experiments. Statistical analysis was performed using SPSS software version 17.0 (SPSS, Inc., Chicago, IL, USA). One-way analysis of variance followed by Tukey post hoc test was conducted for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

RA reduces H2O2-induced cytotoxicity in NHDFs

The present study initially determined the dose range of RA that was not markedly cytotoxic to NHDFs. The cells were treated with increasing concentrations of RA (0–50 µM) for 12 h. Concentrations of RA <50 µM were not were not significantly cytotoxic; therefore, 10–30 µM RA was used for subsequent experiments. To determine if RA affects the viability of NHDFs exposed to H2O2-induced cellular stress, NHDFs were sequentially treated with RA and H2O2, and cytotoxicity was evaluated using a WST-1 assay. As presented in Fig. 1, cell viability decreased to 76% of the control in cells treated with 600 µM H2O2 for 12 h. Pretreatment with 10–30 µM RA for 12 h significantly inhibited H2O2-induced cytotoxicity (Fig. 1B).

RA inhibits H2O2-mediated induction of SA-β-gal activity in NHDFs

H2O2-mediated ROS activates endogenous cellular responses to oxidative stress (30). However, when this defense system collapses, superoxide anion, hydroxyl radical and singlet oxygen production induces cellular senescence and apoptosis (3133). In the present study, H2O2 induced cellular senescence in NHDFs, as demonstrated by an SA-β-gal assay (Fig. 2). Notably, RA markedly inhibited cellular senescence in a concentration-dependent manner in H2O2-exposed NHDFs (Fig. 2), thus suggesting that RA protects NHDFs from H2O2-mediated induction of SA-β-gal activity.

RA inhibits H2O2-induced oxidative stress in NHDFs

Similar to previous studies, the present study investigated whether the protective effects of RA were associated with its oxidative properties (2325). To evaluate the ROS scavenging activity of RA, H2O2 scavenging activity, DCF-DA fluorescence intensity, NRF2 activity and the expression of genes that regulate oxidative stress (HO-1, SOD1 and CAT) were assessed. RA showed an increase in H2O2 scavenging activity and ascorbic acid was used as positive control (Fig. 3A). Although the free radical scavenging activity of RA was less than the positive control (L-ascorbic acid), the half maximal inhibitory concentration of RA was 0.56 mg/ml, and the radical scavenging activity of RA occurred in a dose-dependent manner. DCF fluorescence intensity, which is an indicator of intracellular ROS levels, was evaluated in RA-pretreated NHDFs with or without H2O2 exposure. DCF intensity increased >8-fold in H2O2-exposed cells compared with the nonexposed control group (Fig. 2B). However, this effect was strongly reduced in NHDFs pretreated with either RA or the positive control antioxidant NAC (Fig. 3B). NRF2 serves a key role in the regulation of antioxidant mechanisms by activating the transcription of genes encoding antioxidant enzymes, including HO-1, SOD1 and CAT, via the ARE in the target gene promoter (57). Therefore, the present study analyzed the transcriptional activity of NRF2 using the ARE-luciferase assay. As presented in Fig. 3C, RA markedly enhanced luciferase activity in a concentration-dependent manner, thus suggesting that RA activates NRF2 activity in NHDFs. In addition, the expression levels of the NRF2 target genes, HO-1, SOD1 and CAT, were detected. Notably, RA markedly upregulated the target genes against H2O2-induced oxidative stress (Fig. 3D). These results suggested that RA exerts a protective effect on H2O2-induced oxidative stress in NHDFs via NRF2-associated antioxidant mechanisms.

RA inhibits the H2O2-induced inflammatory response in NHDFs

The mammalian Sir2 ortholog, SIRT1, is a histone deacetylase that targets histones and nonhistone substrates, including NF-κB and p53 (8,9,34). Previous studies have demonstrated that SIRT1 is involved in the regulation of numerous cellular processes, including inflammation, apoptosis and autophagy, and that SIRT1 expression is downregulated in response to oxidative stress (1012,35). Therefore, the present study evaluated the expression levels of SIRT1 and various inflammatory cytokines, including NF-κB, TNF-α and IL-6 in NHDFs. SIRT1 levels were decreased in NHDFs exposed to 600 µM H2O2, whereas RA inhibited this effect in a concentration-dependent manner (Fig. 4A). To determine the effects of H2O2 and RA on NF-κB expression, an NF-κB luciferase reporter stable NIH-3T3 cell line was used. As presented in Fig. 4B, relative luciferase activity in H2O2-exposed NHDFs increased 5.71±0.42-fold compared with the control group (P<0.05). Notably, pretreatment with 20 and 30 µM RA significantly inhibited this effect (P<0.05). To further evaluate the effects of H2O2 and RA on the activity of NF-κB, the expression levels of the NF-κB target genes, TNF-α and IL-6, were determined. H2O2 upregulated TNF-α and IL-6 expression, whereas RA significantly inhibited this effect in a concentration-dependent manner (Fig. 4C). Taken together, these results indicated that RA exerts an anti-inflammatory effect on NHDFs under conditions of excessive oxidative stress.

Discussion

Numerous studies have demonstrated that ROS exist in all living tissues, and are essential for various cellular functions, including epithelial cell proliferation and wound healing (36), and dermal fibroblast migration (37). However, excess ROS can trigger deleterious effects, such as apoptosis and cellular senescence (3). There is a marked decreased in antioxidant levels in aged skin, and treating aged skin with antioxidants, such as resveratrol and vitamin E, can partially rejuvenate aged skin (38). These findings suggested that ROS may be the primary cause for the appearance of aging skin. H2O2 is a type of ROS, which is generated in response to UV light and numerous pollutants (39). The histological alterations in aged skin are markedly visible in the dermis layer, where dermal fibroblasts are the primary cell type (40). In addition, a reduction in CAT activity and an increase in H2O2 levels have been observed in dermal fibroblasts from aged human skin compared with in dermal fibroblasts from younger human skin (41). In a previous study, treatment with exogenous CAT rescued the reduction in intracellular CAT protein activity and the increase in H2O2 levels in aged fibroblasts, and also reduced collagenase matrix metalloproteinase 1 expression (41). These data indicated that suppressing H2O2 generation in dermal fibroblasts may inhibit the skin aging process. Notably, in the present study, H2O2 exposure strongly enhanced cytotoxicity and SA-β-gal activity, which are similar to the effects observed in tissues from older individuals (42). However, RA pretreatment markedly protected NHDFs from H2O2-induced cytotoxicity and SA-β-gal activation. In addition, the H2O2 scavenging capacity of RA was investigated, as was the intracellular H2O2 scavenging capacity of RA in NHDFs. The results demonstrated that RA exhibited substantial H2O2 scavenging activity and inhibited H2O2-induced intracellular ROS production. Taken together, these data suggested that RA may be considered a potential ingredient in anti-aging skin products.

The results of the present study prompted the hypothesis that RA may regulate intracellular antioxidant activity in NHDFs. Although RA has exhibited antioxidant activity in keratinocytes (24,25), there are differences in the oxidative stress response between keratinocytes and dermal fibroblasts (43). To test the hypothesis, ARE luciferase assays and RT-qPCR were conducted to evaluate the expression of the antioxidant-induced NRF2 target genes HO-1, SOD1 and CAT. RA rescued H2O2-mediated inhibition of NRF2 transcriptional activity and the consequential decrease in NRF2 target gene expression. Consistent with these findings, a previous study demonstrated that RA inhibits UVB-induced ROS production and the decrease in protein levels encoded by NRF2 target genes in HaCaT keratinocytes (24). In addition, a previous report demonstrated that the NRF2-inducer tanshinone I exerted a protective effect against UV radiation in human skin cells and reconstructed human skin (44). Furthermore, NRF2 depletion induces damage to the extracellular matrix (45), a hallmark of skin aging (46). Taken together, these findings indicated that RA may inhibit H2O2 via a NRF2-associated antioxidant mechanism in NHDFs.

The present study also demonstrated that RA inhibited the H2O2-induced inflammatory response in NHDFs. Oxidative stress is associated with inflammation in skin tissue (47), and upregulation of the inflammatory response is associated with numerous skin diseases, including atopic dermatitis and psoriasis (48). SIRT1 serves critical roles in apoptosis, senescence, autophagy, gene silencing and inflammation (11,12,48,49). Under excessive ROS conditions, SIRT1 is inactivated and cannot suppress the activity of NF-κB, which is a protein that serves a key role in inflammatory responses by activating the transcription of proinflammatory cytokines, such as TNF-α and IL-6 (16). In the present study, RA inhibited H2O2-mediated SIRT1 downregulation in NHDFs, and NF-κB activity was decreased in RA-treated NHDFs. Furthermore, RA significantly downregulated the expression levels of TNF-α and IL-6 in a dose-dependent manner. NF-κB activation is regarded as a potential biomarker of aging (14); an increase in NF-κB activity is correlated with tissue aging and age-associated degenerative diseases (15).

In conclusion, the findings of the present study suggested that RA may inhibit senescence and inflammatory responses in NHDFs, which are effects associated with aging skin. To gain insight into the effects of RA on human skin, further studies should focus on the efficacy of RA in reversing the signs of aging skin and evaluate the precise molecular mechanisms by which RA mediates this effect in vivo.

Acknowledgements

The present study was supported by a grant from the Ministry of Trade, Industry and Energy (MOTIE), Korea Institute for Advancement of Technology (KIAT) through the Encouragement Program for The Industries of Economic Cooperation Region (grant no. R0003962) and a grant from the Marine Biotechnology Program (grant no. 20150184) funded by the Ministry of Oceans and Fishers, Republic of Korea.

References

1 

Poljsak B, Dahmane RG and Godić A: Intrinsic skin aging: The role of oxidative stress. Acta Dermatovenerol Alp Pannonica Adriat. 21:33–36. 2012.PubMed/NCBI

2 

Ganceviciene R, Liakou AI, Theodoridis A, Makrantonaki E and Zouboulis CC: Skin anti-aging strategies. Dermatoendocrinol. 4:308–319. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Davalli P, Mitic T, Caporali A, Lauriola A and D'Arca D: ROS, cell senescence, and novel molecular mechanisms in aging and age-related diseases. Oxid Med Cell Longev. 2016:35651272016. View Article : Google Scholar : PubMed/NCBI

4 

Kansanen E, Kuosmanen SM, Leinonen H and Levonen AL: The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 1:45–49. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Kensler TW, Wakabayashi N and Biswal S: Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 47:89–116. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al: An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 236:313–322. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Buendia I, Michalska P, Navarro E, Gameiro I, Egea J and Leon R: Nrf2-ARE pathway: An emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacol Ther. 157:84–104. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L and Gu W: Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell. 107:137–148. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, Guarente L and Weinberg RA: hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 107:149–159. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Wu A, Ying Z and Gomez-Pinilla F: Oxidative stress modulates Sir2alpha in rat hippocampus and cerebral cortex. Eur J Neurosci. 23:2573–2580. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Moon MH, Jeong JK, Lee YJ, Seol JW, Jackson CJ and Park SY: SIRT1, a class III histone deacetylase, regulates TNF-α-induced inflammation in human chondrocytes. Osteoarthritis Cartilage. 21:470–480. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Takayama K, Ishida K, Matsushita T, Fujita N, Hayashi S, Sasaki K, Tei K, Kubo S, Matsumoto T and Fujioka H: SIRT1 regulation of apoptosis of human chondrocytes. Arthritis Rheum. 60:2731–2740. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Salminen A, Kauppinen A, Suuronen T and Kaarniranta K: SIRT1 longevity factor suppresses NF-kappaB-driven immune responses: Regulation of aging via NF-kappaB acetylation? Bioessays. 30:939–942. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Balistreri CR, Candore G, Accardi G, Colonna-Romano G and Lio D: NF-κB pathway activators as potential ageing biomarkers: Targets for new therapeutic strategies. Immun Ageing. 10:242013. View Article : Google Scholar : PubMed/NCBI

15 

Tilstra JS, Clauson CL, Niedernhofer LJ and Robbins PD: NF-κB in Aging and Disease. Aging Dis. 2:449–465. 2011.PubMed/NCBI

16 

Tak PP and Firestein GS: NF-kappaB: A key role in inflammatory diseases. J Clin Invest. 107:7–11. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Vladimir-Knezevic S, Blazekovic B, Kindl M, Vladic J, Lower-Nedza AD and Brantner AH: Acetylcholinesterase inhibitory, antioxidant and phytochemical properties of selected medicinal plants of the Lamiaceae family. Molecules. 19:767–782. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Jang AH, Kim TH, Kim GD, Kim JE, Kim HJ, Kim SS, Jin YH, Park YS and Park CS: Rosmarinic acid attenuates 2,4-dinitrofluorobenzene-induced atopic dermatitis in NC/Nga mice. Int Immunopharmacol. 11:1271–1277. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Karthik D, Viswanathan P and Anuradha CV: Administration of rosmarinic acid reduces cardiopathology and blood pressure through inhibition of p22phox NADPH oxidase in fructose-fed hypertensive rats. J Cardiovasc Pharmacol. 58:514–521. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Kim GD, Park YS, Jin YH and Park CS: Production and applications of rosmarinic acid and structurally related compounds. Appl Microbiol Biotechnol. 99:2083–2092. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Khojasteh A, Mirjalili MH, Hidalgo D, Corchete P and Palazon J: New trends in biotechnological production of rosmarinic acid. Biotechnol Lett. 36:2393–2406. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Sanchez-Campillo M, Gabaldon JA, Castillo J, Benavente-García O, Del Baño MJ, Alcaraz M, Vicente V, Alvarez N and Lozano JA: Rosmarinic acid, a photo-protective agent against UV and other ionizing radiations. Food Chem Toxicol. 47:386–392. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Yoo SM and Kang JR: Antioxidation effects of rosmarinic acid on human skin melanoma cells treated with hydrogen peroxide. J Korean Soc Appl Bi. 52:247–251. 2009. View Article : Google Scholar

24 

Fernando PM, Piao MJ, Kang KA, Ryu YS, Hewage SR, Chae SW and Hyun JW: Rosmarinic acid attenuates cell damage against UVB radiation-induced oxidative stress via enhancing antioxidant effects in human HaCaT cells. Biomol Ther (Seoul). 24:75–84. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Vostálová J, Zdarilová A and Svobodová A: Prunella vulgaris extract and rosmarinic acid prevent UVB-induced DNA damage and oxidative stress in HaCaT keratinocytes. Arch Dermatol Res. 302:171–181. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Oyedemi SO, Bradley G and Afolayan AJ: In-vitro and -vivo antioxidant activities of aqueous extract of Strychnos henningsii Gilg. Afr J Pharm Pharmaco. 4:70–78. 2010.

27 

Martinez-Outschoorn UE, Trimmer C, Lin Z, Whitaker-Menezes D, Chiavarina B, Zhou J, Wang C, Pavlides S, Martinez-Cantarin MP, Capozza F, et al: Autophagy in cancer associated fibroblasts promotes tumor cell survival: Role of hypoxia, HIF1 induction and NFκB activation in the tumor stromal microenvironment. Cell Cycle. 9:3515–3533. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Debacq-Chainiaux F, Erusalimsky JD, Campisi J and Toussaint O: Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc. 4:1798–1806. 2009. View Article : Google Scholar : PubMed/NCBI

29 

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 : PubMed/NCBI

30 

Martindale JL and Holbrook NJ: Cellular response to oxidative stress: Signaling for suicide and survival. J Cell Physiol. 192:1–15. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Korycka-Dahl MB and Richardson T: Activated oxygen species and oxidation of food constituents. CRC Crit Rev Food Sci Nutr. 10:209–241. 1978. View Article : Google Scholar : PubMed/NCBI

32 

Halliwell B and Gutteridge JM: Oxygen free radicals and iron in relation to biology and medicine: Some problems and concepts. Arch Biochem Biophys. 246:501–514. 1986. View Article : Google Scholar : PubMed/NCBI

33 

Simon HU, Haj-Yehia A and Levi-Schaffer F: Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 5:415–418. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA and Mayo MW: Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 23:2369–2380. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Ou X, Lee MR, Huang X, Messina-Graham S and Broxmeyer HE: SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells. 32:1183–1194. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Huo Y, Qiu WY, Pan Q, Yao YF, Xing K and Lou MF: Reactive oxygen species (ROS) are essential mediators in epidermal growth factor (EGF)-stimulated corneal epithelial cell proliferation, adhesion, migration, and wound healing. Exp Eye Res. 89:876–886. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Shi HX, Cheng Y, Ye JJ, Cai P, Zhang J, Li R, Yang Y, Wang Z, Zhang H, Lin C, et al: bFGF promotes the migration of human dermal fibroblasts under diabetic conditions through reactive oxygen species production via the PI3K/Akt-Rac1-JNK pathways. Int J Biol Sci. 11:845–859. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Farris P, Yatskayer M, Chen N, Krol Y and Oresajo C: Evaluation of efficacy and tolerance of a nighttime topical antioxidant containing resveratrol, baicalin, and vitamin e for treatment of mild to moderately photodamaged skin. J Drugs Dermatol. 13:1467–1472. 2014.PubMed/NCBI

39 

Peus D, Vasa RA, Meves A, Pott M, Beyerle A, Squillace K and Pittelkow MR: H2O2 is an important mediator of UVB-induced EGF-receptor phosphorylation in cultured keratinocytes. J Invest Dermatol. 110:966–971. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Gilchrest BA: Skin aging and photoaging: An overview. J Am Acad Dermatol. 21:610–613. 1989. View Article : Google Scholar : PubMed/NCBI

41 

Shin MH, Rhie GE, Kim YK, Park CH, Cho KH, Kim KH, Eun HC and Chung JH: H2O2 accumulation by catalase reduction changes MAP kinase signaling in aged human skin in vivo. J Invest Dermatol. 125:221–229. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC, Kleijer WJ, DiMaio D and Hwang ES: Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell. 5:187–195. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Marionnet C, Pierrard C, Lejeune F, Sok J, Thomas M and Bernerd F: Different oxidative stress response in keratinocytes and fibroblasts of reconstructed skin exposed to non extreme daily-ultraviolet radiation. PLoS One. 5:e120592010. View Article : Google Scholar : PubMed/NCBI

44 

Tao S, Justiniano R, Zhang DD and Wondrak GT: The Nrf2-inducers tanshinone I and dihydrotanshinone protect human skin cells and reconstructed human skin against solar simulated UV. Redox Biol. 1:532–541. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Saw CL, Yang AY, Huang MT, Liu Y, Lee JH, Khor TO, Su ZY, Shu L, Lu Y, Conney AH and Kong AN: Nrf2 null enhances UVB-induced skin inflammation and extracellular matrix damages. Cell Biosci. 4:392014. View Article : Google Scholar : PubMed/NCBI

46 

Panich U, Sittithumcharee G, Rathviboon N and Jirawatnotai S: Ultraviolet radiation-induced skin aging: The role of DNA damage and oxidative stress in epidermal stem cell damage mediated skin aging. Stem Cells Int. 2016:73706422016. View Article : Google Scholar : PubMed/NCBI

47 

Wagener FA, Carels CE and Lundvig DM: Targeting the redox balance in inflammatory skin conditions. Int J Mol Sci. 14:9126–9167. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Rahman I, Kinnula VL, Gorbunova V and Yao H: SIRT1 as a therapeutic target in inflammaging of the pulmonary disease. Prev Med. 54 Suppl:S20–S28. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Yao H and Rahman I: Perspectives on translational and therapeutic aspects of SIRT1 in inflammaging and senescence. Biochem Pharmacol. 84:1332–1339. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hahn HJ, Kim KB, An IS, Ahn KJ and Han HJ: Protective effects of rosmarinic acid against hydrogen peroxide‑induced cellular senescence and the inflammatory response in normal human dermal fibroblasts. Mol Med Rep 16: 9763-9769, 2017
APA
Hahn, H.J., Kim, K.B., An, I., Ahn, K.J., & Han, H.J. (2017). Protective effects of rosmarinic acid against hydrogen peroxide‑induced cellular senescence and the inflammatory response in normal human dermal fibroblasts. Molecular Medicine Reports, 16, 9763-9769. https://doi.org/10.3892/mmr.2017.7804
MLA
Hahn, H. J., Kim, K. B., An, I., Ahn, K. J., Han, H. J."Protective effects of rosmarinic acid against hydrogen peroxide‑induced cellular senescence and the inflammatory response in normal human dermal fibroblasts". Molecular Medicine Reports 16.6 (2017): 9763-9769.
Chicago
Hahn, H. J., Kim, K. B., An, I., Ahn, K. J., Han, H. J."Protective effects of rosmarinic acid against hydrogen peroxide‑induced cellular senescence and the inflammatory response in normal human dermal fibroblasts". Molecular Medicine Reports 16, no. 6 (2017): 9763-9769. https://doi.org/10.3892/mmr.2017.7804