DOI QR코드

DOI QR Code

A new role for the ginsenoside RG3 in antiaging via mitochondria function in ultraviolet-irradiated human dermal fibroblasts

  • Lee, Hyunji (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Hong, Youngeun (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Tran, Quangdon (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Cho, Hyeonjeong (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Kim, Minhee (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Kim, Chaeyeong (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Kwon, So Hee (Department of Pharmacy, College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University) ;
  • Park, SungJin (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Park, Jongsun (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Park, Jisoo (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University)
  • Received : 2018.04.05
  • Accepted : 2018.07.09
  • Published : 2019.07.15

Abstract

Background: The efficacy of ginseng, the representative product of Korea, and its chemical effects have been well investigated. The ginsenoside RG3 has been reported to exhibit apoptotic, anticancer, and antidepressant-like effects. Methods: In this report, the putative effect of RG3 on several cellular function including cell survival, differentiation, development and aging process were evaluated by monitoring each specific marker. Also, mitochondrial morphology and function were investigated in ultraviolet (UV)-irradiated normal human dermal fibroblast cells. Results: RG3 treatment increased the expression of extracellular matrix proteins, growth-associated immediate-early genes, and cell proliferation genes in UV-irradiated normal human dermal fibroblast cells. And, RG3 also resulted in enhanced expression of antioxidant proteins such as nuclear factor erythroid 2-related factor-2 and heme oxygenase-1. In addition, RG3 affects the morphology of UV-induced mitochondria and plays a role in protecting mitochondrial dysfunction. Conclusioin: RG3 restores mitochondrial adenosine triphosphate (ATP) and membrane potential via its antioxidant effects in skin cells damaged by UV irradiation, leading to an increase in proteins linked with the extracellular matrix, cell proliferation, and antioxidant activity.

Keywords

1. Introduction

Ultraviolet (UV) irradiation is well known as a main factor of skin aging [1-3]. The primary features of skin aging in humans include a decline in fibrous tissue, slow cellular regeneration, and a reduction of the vascular and glandular network [4]. The extracellular matrix (ECM) components of the skin must be well balanced to provide structurally functional support [5].

Collagen is the basic protein of structure in the extracellular space of various types of animal connective tissues and constitutes approximately 25-35% of all protein content in the body [6]. Matrix metalloproteinases (MMPs) degrade ECM connective tissue [7].

In addition to UV irradiation, other reactive oxygen species (ROS) and hydrogen peroxide (H2O2) are involved in the pathogenesis of several skin conditions, including aging, wrinkles, photosensitivity, and malignancy [8].

Commonly used in the pharmaceutical and cosmetic industries, plant extracts with antioxidant properties decrease the production of ROS. Various traditional plant extracts have well-known properties, including medicinal functions, pharmacological activities, and skin-protective effects [9,10]. The efficacy of ginseng, the representative product of Korea, and its chemical effects have been well investigated. The ginsenoside RG3 has been reported to exhibit apoptotic [11,12], anticancer [13,14], antiinflammatory {Cheng, 2016 #17], and antidepressant-like effects [15].

2. Materials and methods

2.1. RG3 material

The ginsenoside RG3 (>95% purity) was purchased from Korean Tobacco and Ginseng, Ltd. (Deajon, Korea).

2.2. Cell culture and cell viability

Cell culture and the cell viability assay were performed as previously described [16].

2.3. Wound healing assay

For the cell migration assay, monolayers were carefully scratched using a 10-μL pipette tip. After 24 hr of UV irradiation (40 J/m2) and treatment with UV crosslinker (Vilber Lourmat, Collégien, France) for 3 min, cells were untreated or treated with 1, 5, or 10 μM RG3 extract for 24 h. Wounded areas were then photographed.

2.4. Western blot analysis and antibodies

Western blotting was performed as previously described [16,17]. Anticollagen (Abcam, Cambridge, UK), anti-dynamin-related protein 1 (DRP1), and anti-GTPase optic atrophy 1 (OPA1) antibodies were purchased from BD Biosciences (San Jose, CA, USA). Anti-actin antibody was obtained from Sigma-Aldrich (St. Louis, MO, USA), anti-phosphorylated DRP1 [pDRP1; phosphorylated serine 616 (pS616) or pS637] was purchased from Cell Signaling Technology (Boston, MA, USA), and anti-elastin, anti-MMP-3, anti-extracellular signaleregulated kinase (ERK), anti-translocase of mitochondrial outer-membrane 40 (TOM40), anti-mitofusin (MFN), and antimitochondrial fission protein 1 (FIS1) antibodies were obtained from Santa Cruz Biotechnology (Dallas, TX, USA). Horseradish peroxidaseeconjugated anti-mouse or anti-rabbit IgG secondary antibodies were obtained from Koma Biotech (Seoul, Korea).

2.5. Quantitative polymerase chain reaction

Quantitative polymerase chain reaction was performed as previously described [16].

2.6. ROS assay

Normal human dermal fibroblast (NHDF) cells were plated and incubated at a density of 1.0 ×106 cells/well in six-well culture plates for 24 h. The following day, cells were UV irradiated with 40 J/m2 followed by treatment with UV crosslinker (Vilber Lourmat) for 3 min and either untreated or treated with RG3 extract for 24 h. Cells were washed, stained with 5 μM 2',7'-dichlorodihydrofluorescein diacetate (CM-H2DCFDA; Life Technologies, Carlsbad, CA, USA) and 20 mM dihydroethidium (DHE) in Hank’s balanced salt solution and incubated at 37°C for 30 min. FACSCalibur flow cytometer (BD Biosciences) was used to measure H2O2 activity using an emission filter at 532 nm. At least 10,000 cells were analyzed in three independent experiments.

2.7. Mitochondrial fraction

NHDF cells were washed with phosphate buffered saline (PBS) and resuspended in mitochondrial fraction buffer (20 mM 4-(2-Hydroxyethyl)piperazine-1-ethanesulfonic acid, N-(2-Hydroxyethyl) piperazine-N'-(2-ethanesulfonic acid) [HEPES], pH 8.0,10 mM KCl,1.5 mM MgCl2, 1 mM ethylenedinitrilo)tetraacetic acid (EDTA), 250 mM sucrose, 1 mM phenylmethane sulfonyl fluoride, 10 g/mL leupeptin, 10 g/mL aprotinin, and 0.2 mM sodium orthovanadate) for 30 min on ice, and homogenized. Unbroken cells and nuclei were pelleted by centrifugation at 1500× g for 10 min. The supernatant was centrifuged at 10,000×g for 30 min at 4°C and transferred to a new tube as the postmitochondrial fraction. Then, the supernatant was centrifuged at 10,000× g for 1 h at 4°C, and the resulting supernatant was used as the cytosolic fraction. The pellet containing the postmitochondrial fraction was washed with 500 μL of mitochondrial fraction buffer and used to isolate the mitochondrial fraction. Isolated mitochondria were treated with 2M sodium chloride, 100 mM sodium carbonate (pH 11.2), or 1% Triton X-100 for 30 min. The samples were ultracentrifuged to separate the supernatant (S) and precipitate (P) fractions, which were used to analyze tricarboxylic acid activity.

2.8. Confocal imaging analysis

NHDF cells were grown on glass coverslips to 50e70% confluency and transfected with pDsRed2-Mito using jetPEI reagents (polyplus transfection, NY, USA). After 48 h, cells were fixed in 4% paraformaldehyde at room temperature for 10 min and permeabilized with 0.2% Triton X-100 for 5 min at room temperature. Then, cells were incubated in blocking buffer containing 5% bovine serum albumin (Sigma-Aldrich) in 1×Tris-buffered saline for 1 h at 37°C. The rabbit polyclonal anti-cytochrome-c antibody was diluted 200-fold for use as a primary antibody and incubated overnight. Fluorescein isothiocyanateeconjugated anti-mouse antibody (BD Biosciences, Franklin Lakes, NJ, USA) was used as the secondary antibody. After appropriate rinsing, coverslips were mounted with Vectashield (Vector Laboratories, Burlingame, CA, USA) and visualized using a Leica confocal microscope (Leica Microsystems, Wetzlar, Germany].

2.9. Measurement of ATP levels in subcellular compartments

Mitochondrial adenosine triphosphate (ATP) was measured using a firefly luciferase construct conjugated with a COX8 mitochondrial targeting sequence (pcDNA3.1/Zeo-CHA-COX8-Luc) as previously described [18]. Briefly, light emission was measured in a luminometer at 5-s intervals until the maximum value of luminescence was reached. To normalize for the variability of luciferase expression in transfected cells, the relative luminescence values in each cellular compartment were expressed as a ratio to the total potential luminescence measured in equal amounts of the same lysed cells with a luciferase assay kit (Promega, Madison, WI, USA) in the presence of excess ATP (final concentration, 0.5 mmol/L).

2.10. Measurement of mitochondrial membrane potential (∆ψm)

NHDF cells were irradiated with 40 J/m2 UV followed by treatment with UV crosslinker (Vilber Lourmat) for 3 min and either untreated or treated with 10 μM of RG3 extract for 24 h or 30 μM carbonyl cyanide m-chlorophenyl hydrazone (CCCP) for 8 h as a control. Cells were stained with 1 μM rhodamine-123 for 60 min to measure the mitochondrial transmembrane potential using flow cytometry. A minimum of 10,000 cells per sample were analyzed.

2.11. Statistical analysis

Statistical analysis was performed as previously described [16].

3. Results

3.1. Cell viability and wound healing of NHDF cells treated with RG3 extract

NHDF cells were treated with 1, 5, and 10 μM of RG3 extract for 24 hr. Compared with untreated cells, treatment with the RG3 extract did not exhibit cytotoxicity (Fig. 1A). To assess the antiapoptotic effect of RG3, NHDF cells were induced to undergo apoptosis by exposure to 60 J/m2 of UVC irradiation. The apoptosis marker caspase 3 was cleaved in a time-dependent manner (Fig. 1B). When confluent monolayers of cells were treated with RG3 for 24 hr after UV irradiation in a wound healing assay, cells closed readily when treated with doses from 1 to 10 μM in a dosedependent manner (Fig. 1C).

Fig. 1. Cell viability assay and cell wound healing assay. (A) RG3 extract did not result in cytotoxicity at 1, 5, and 10 μg/mL by EZ-Cyto assay with NHDF cells. (B) Induction of apoptotic cell death was measured by Western blot analysis with anti-caspase 3 antibodies after UV irradiation of 40 J/m2 in NHDF cells. (C) Cell wound healing analysis was applied to UV-irradiated cells for 24 hours after scratching. The UV-irradiated NHDF cells were further treated with RG3 extract in a dose-dependent manner. NHDF, normal human dermal fibroblast; UV, ultraviolet.

3.2. Cell differentiation, development, and protective effects of RG3 after UV irradiation in NHDF cells

Protein kinase B (PKB)/AKT plays a fundamental role in signaling pathways that regulate a variety of cellular functions, such as cell survival, growth, nutrient metabolism, proliferation, and apoptosis.

Phosphorylation of PKB/AKT at Ser473 and Thr308 fully activates PKB/AKT [19]. Exposure of mouse epidermal JB6 Cl 41 cells to UV irradiation led to phosphorylation of PKB/AKT at Ser473 and Thr308 in a time-dependent manner. UV irradiation induced PKB/AKT phosphorylation of epidermal cells, indicating a correlation between the response to UV and PKB/AKT [20]. Activated ERKs translocate to the nucleus and promote changes in gene expression, growth, differentiation, or mitosis [21]. Mammalian cells respond quickly when exposed to UV, stimulating various factors, which ultimately results in the activation of transcription factors and proteins such as mitogen-activated protein kinases (MAPKs) and ERKs. MAPKs are activated by phosphorylation of specific tyrosine and theronine residues [22,23].

We also investigated the effect of UV irradiation on the phosphorylation and activity of PKB/AKT (S473) and ERK (T202 and Y204). Total protein levels of ERK were upregulated in RG3-treated cells after UV irradiation (Figs. 2A, 2B).

Protein extracts of NHDF cells either untreated or treated with RG3 extract for 24 h after UV irradiation were analyzed by Western blot. It has previously been reported that in human fibroblasts, type I collagen gene expression was regulated by the ERK signaling pathway [24]. Our data showed that the ERK protein level was dramatically increased in a dose-dependent manner (Fig. 2A). Expression of phosphorylated ERK (T44/T42) and PKB (S473) was significantly increased in a dose-dependent manner in UV-induced NHDF cells (Fig. 2A). UV irradiation is known to affect immuneregulatory factors and signal transduction in the skin. More specifically, UV irradiation increases the expression of several growthassociated immediate-early genes such as c-Jun [25,26]. Tumor necrosis factor (TNF)-α, which was the first cytokine linked to immunosuppression and skin carcinogenesis after UV irradiation, increases the production of TNF-a in keratinocytes during UV irradiation and contributes to the induction of skin cancer [27-29]. Growth factor receptor, Src, and Ras-Raf-MEK-MAPK signaling pathways are also important after UV exposure [30-34]. Rapid nuclear translocation of epidermal growth factor receptor (EGFR) was observed in UV-irradiated human primary and HaCaT keratinocytes. UV irradiation induces nuclear translocation of the EGFR in human keratinocytes [35]. It has also been reported that structural changes were induced by UVB light in the epidermal growth factor-binding site [36]. Our data indicate that the expression of TNF receptor 1, EGFR, c-Jun, and pp38 was decreased in a dosedependent manner in UV-induced NHDF cells (Figs. 2C, 2D).

Fig. 2. Cell differentiation, development, and proliferation protein expression by Western blot. (A, C) Total protein extracted from untreated or RG3 extract-treated cells was analyzed by Western blot with anti-extracellular signal-regulated kinase (ERK), anti-phosphorylated ERK2, anti-protein kinase B (PKB), anti-phosphorylated PKB, anti-tumor necrosis factor receptor 1 (TNFR1), anti-epidermal growth factor receptor (EGFR), anti-c-JUN, and anti-phosphorylated-p38 antibodies. (B, D) Anti-ERK, anti-pERK, anti-PKB, anti-pPKB, anti-TNFR1, anti-EGFR, anti-c-JUN, and anti-pp38 were quantified by Western blotting, as described in the Materials and methods section. The values are means ± SD of three independent experiments. Significant difference from negative control (Non-UV irradiated): *, p < 0.05 and **, p < 0.01. Significant difference from positive control (UV irradiated): #, p < 0.05 and ##, p < 0.01. pERK, phosphorylated ERK; SD, standard deviation; UV, ultraviolet.

3.3. RG3 enhanced the expression of ECM proteins in UV-irradiated NHDF cells

The general composition of the ECM varies between multicellular structures due to cell adhesion, intercellular communication, and differentiation [37,38]. Development, tissue repair, morphogenesis, and remodeling are characterized by degradation of the ECM. Normal physiological conditions regulate decomposition of the ECM; however, disability of this process can result in disease such as chronic inflammation and cancer [39].

Collagen is the most abundant fibrous protein within the interstitial ECM and as the main structural elements of ECM provide tensile strength, control cell adhesion, support chemotaxis and migration, and induce tissue development [40,41]. Collagen associates with elastin, and stretching is limited by close association with collagen fibers [42]. Elastin is a highly elastic ECM protein in connective tissue that helps maintain tissue integrity after stretching and contraction, especially when the skin is pinched or pressed. The elastin of the tissue is combined with the fine fibers to form elastic fibers and in adult tissues constitutes the main component of elastic fibers [43].

In humans, alpha-1 type I collagen (COL1A1) is encoded by the COL1A1 gene, which encodes a major component of type I collagen, and includes fibrin collagen found in connective tissues, including cartilage [44]. After UV irradiation, skin is damaged. Infiltrating macrophages move to the site of damage and release interferon-γ, which binds to receptors in sun-damaged melanocytes to avoid detection by the immune system [45]. It is well known that interferon-γ inhibits COL1A1 expression in fibroblasts, principally at the transcriptional level [46].

We evaluated the mRNA levels of ECM proteins in RG3-treated cells after UV irradiation. Our data showed that the mRNA levels of collagen, elastin, and COL1A1 were increased in a dosedependent manner, whereas that of MMP-2 was not (Fig. 3A). Protein extracts of untreated or RG3-treated cells after UV irradiation were analyzed by Western blotting. After UV exposure, the basal expression level of collagen and elastin was decreased dramatically, but RG3-treated cells showed increased collagen and elastin levels compared with untreated controls. The protein content of MMP-3 was increased compared to that of the normal state during UV irradiation, but the expression of MMP-3 decreased in proportion to the concentration of RG3 after UV irradiation (Figs. 3B, 3C).

It has been reported that the ERK signaling pathway regulates type I collagen gene expression in human fibroblasts [47]. Our data showed that the ERK protein level was significantly increased and that expression of the interstitial collagenase enzyme MMP-3 was significantly downregulated in a dose-dependent manner (Figs. 2A and 3A, 3B). These results suggested that the antiaging effects of RG3 treatment in UV-irradiated cells are associated with the transcriptional and translational regulation of molecules involved in the ECM.

Fig. 3. Extracellular matrix mRNA and protein expression. (A) mRNA level of ECM genes in RG3-treated cell on UV irradiation. (B) Protein level of ECM in RG3-treated cell on UV irradiation. (C) Collagen, elastin, MMP-3, and GAPDH in the immunoprecipitation were quantified by Western blot analyses, as described in the Materials and methods section. Bar heights are means ± SD of three independent experiments. Significant difference from negative control (Non-UV irradiated): *, p < 0.05 and **, p < 0.01. Significant difference from positive control (UV irradiated): #, p < 0.05 and ##, p < 0.01. COL1A1, alpha-1 type I collagen; ECM, extracellular matrix; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; MMP, matrix metalloproteinase; SD, standard deviation; UV, ultraviolet.

3.4. Treatment with RG3 extract inhibited apoptosis after UV exposure in NHDF cells

UVA/UVB light stimulates the production of ROS such as O2•-, H2O2, and 1O2 in the skin [48-50]. The production of ROS occurs during physiological processes such as aging, metabolism, and apoptosis, as well as in a number of pathological conditions. Environmental stresses can lead to harmful oxidative reactions, and ROS are abolished via enzymatic as well as nonenzymatic antioxidative mechanisms [33]. Antioxidants are well known as a systemic light protectant against UVAand UVB-induced skin damage [51]. For the removal of ROS, every organism has a complex defense system consisting of both antioxidants and antioxidant enzymes [52].

Exposure to UV or H2O2increases cell toxicity. In particular, H2O2exposure in muscle cells may increase cytotoxic inflammation and increase H2O2in atherosclerosis or chronic inflammatory disease [53]. ROS production has been shown to be increased in NHDF cells after UV irradiation [16]. In this study, fluorescence-activated cell sorting revealed that RG3-treated cells exhibit reduced production of ROS compared with untreated cells. The levels of H2O2 were measured using the cell-permeable compound CM-H2DCFDA (Fig. 4A). UV irradiation increased the levels of H2O2, whereas ROS levels were reduced after treatment with RG3. These results indicated that the RG3 protects dermal fibroblasts from UV irradiation by reducing the production of ROS.

It has previously been reported that RG3 inhibits the release of cytochrome-c from depleted serum into the cytoplasm, indicating that mitochondrial-dependent caspase activity is inhibited by RG3 [54]. Cytochrome-c in total cell lysate and the release of cytochrome-c from mitochondria into the cytosol were increased the release of after UV irradiation. We also showed that cytochrome-c into the cytosol was decreased after treatment with RG3 (Fig. 4B), suggesting that apoptosis of UV-exposed skin cells is inhibited by RG3 treatment.

Fig. 4. RG3 inhibits ROS through antioxidant effect. (A) ROS measurement of H2O2 using 2' , 7' -dichlorodehydrofluorosine diacetate (CM-H2DCFDA). (B) The expression level of cytochrome-c after the isolation of mitochondria and cytosols in RG3-treated cell line after UV irradiation. (C, D) qPCR analysis was performed using total RNA from UV-exposed RG3-treated samples. NRF2, HO-1, and β-actin genes were amplified by qPCR using corresponding primers. (E) Protein level of NRF2 and HO-1 from UV-exposed RG3-treated samples by Western blot. (F) Graphical representation. Significant difference from negative control (Non-UV irradiated): *, p < 0.05 and **, p < 0.01. Significant difference from positive control (UV irradiated): #, p < 0.05 and ##, p < 0.01. HO-1, heme oxygenase-1; NRF2, nuclear factor erythroid 2erelated factor-2; qPCR, quantitative polymerase chain reaction; ROS, reactive oxygen species; SD, standard deviation; UV, ultraviolet.

Nuclear factor erythroid 2-related factor-2 (NRF2) increases detoxification pathways and antioxidant potential and protects cardiac fibroblasts and cardiomyocytes against oxidative stress [55,56]. NRF2 also activates the antioxidant response against oxidative or electrophilic stress [57]. Thus, NRF2 is a major player in antioxidant and antiinflammatory response signaling pathways [58]. UVA can cause more damage to cells deficient in antioxidant defense, and low levels of UVA lead to the antioxidant defense regulated by NRF2 [59]. NRF2 regulates cellular resistance to oxidants, and NRF2 knockout mice have been shown to exhibit increased susceptibility to diseases related to chemical toxicity and oxidative pathology [60]. In UVA irradiation in human corneal endothelial cells, antioxidant protection is controlled by increased transcription and translation of NRF2 [59]. NRF2 has also been shown to increase heme oxygenase (HO)-1 after UVA (320-380 nm) irradiation of human skin fibroblasts to control antioxidant activity [61].

Examination of HO-1 and NRF2 mRNA and protein levels in the presence or absence of RG3 treatment after UV exposure revealed that mRNA and protein expression increased in a dose-dependent manner (Figs. 4C-4F). Our results suggest that treatment with RG3 increases the expression of NRF2, which upregulates the expression of HO-1 after inflammation and oxidative stress (Figs. 4C-4F).

3.5. RG3-induced changes in mitochondrial activity and morphology after UV exposure

Mitochondrial quality is maintained by performing qualitative control through a dynamic interconnected network that is continuously dividing and fusing over short periods of time. The proteins that mediate mitochondrial division are DRP1, mitochondrial fission factor (MFF), and FIS1, which are large GTPases, and mitochondrial fusion proteins (MFN1, MFN2), and OPA1, which are GTPases involved in mitochondrial fusion [62-64]. Phosphorylation of DRP1 on Ser616 promotes mitochondrial fission, whereas Ser637 phosphorylation of DRP1 inhibits mitochondrial fission [65]. Mitochondria have been shown to execute a variety of roles in processes such as the transduction of metabolic and stress signals [66] and the production of free radicals such as ROS [67]. When UV light is applied to keratinocytes [68], H2O2 accumulates in mitochondria, causing mutations in mitochondrial DNA, which results in skin aging and skin cancer [69]. Mitochondrial DNA is also known as a biomarker that can present genetic damage by ultraviolet light in the skin of animals and humans [70]. Mitochondria are extremely dynamic organelles whose morphology is regulated by fusion and fission [71,72]. Our data have shown that mitochondrial proteins related to fusion, such as OPA1, MFN2, andpDRP1 (S637), were increased and that one of the mitochondrial proteins related to fission pDRP1 (S616) was decreased, but other fission proteins, such as FIS1 and MFF, were unchanged (Figs. 5A, 5B). Confocal imaging showed that after UV exposure, mitochondria fragment into even smaller units compared with the absence of UV exposure. Treatment with RG3 (10 μM) rescued mitochondrial morphology in UV-exposed NHDF cells (Fig. 5C). Similar to the reduction in mitochondrial membrane potential induced by UV irradiation in keratinocytes [73], the UV irradiationeinduced decrease in mitochondrial membrane potential in NHDF cells was increased after RG3 treatment (Fig. 5D). Following UV exposure, mitochondrial membrane potential and mitochondrial ATP were significantly elevated in RG3-treated cells compared with control cells (Figs. 5D, 5E).

Fig. 5. Changes in mitochondrial activity and morphology by RG3 treatment after UV exposure. (A) The expression level of fusion and fission proteins in RG3-treated cell line after UV irradiation. (B) Graphical representation. (C) Image of MitoRed staining from UV-exposed RG3-treated samples. (D) Mitochondrial membrane potential (∆ψm; mitochondrial respiratory capacity) from UV-exposed RG3-treated samples. (E) ATP of mitochondria from UV-exposed RG3-treated samples. Significant difference from negative control (Non-UV irradiated): *, p < 0.05 and **, p < 0.01. Significant difference from positive control (UV irradiated): #, p < 0.05 and ##, p < 0.01. DRP1. dynamin-related protein 1; FIS1, mitochondrial fission protein 1; MFN2, mitofusin-2; UV, ultraviolet.

4. Discussion

RG3 plants have been investigated for different pharmacological properties; however, no study of the mitochondria of human fibroblast cells after UV damage has been performed. Here, we analyzed several beneficial activities, including antiaging, antioxidant, and recovered mitochondrial functional properties of UV-exposed NHDF cells treated with RG3 extract. Skin is constantly exposed to oxidative stress induced by the production of ROS, and research has focused on antioxidants to combat the pathological conditions caused by oxidative stress [32,74].

Pretreatment of UV-exposed NHFB cells with RG3 extract inhibited the detrimental effects of oxidative stress, as demonstrated by the decrease in ROS generation and increase in cell growth, survival, and proliferation and signaling pathways, including ERK-and AKT/PKB-dependent pathways. Treatment of UV-exposed NHFB cells with RG3 extract also resulted in increased levels of ECM proteins such as collagen and elastin but decreased the function of inhibitory collagenases, such as MMP-2 and MMP-3 (Fig. 3).

Cytochrome-c, a proapoptotic factor released from mitochondria after UV irradiation, was reduced after treatment with RG3. This suggests that UV irradiation induces apoptosis and that the antioxidant activity of RG3 suppresses the release of cytochrome-c from mitochondria and inhibits apoptosis. The antioxidant activity of RG3 in mitochondria transformed mitochondria fragmented by UV exposure into the tubular form and increased the expression of fusion proteins (Figs. 4B, 4C and 5A, 5B).

RG3 restores mitochondrial ATP and membrane potential via its antioxidant effects in skin cells damaged by UV irradiation, leading to an increase in proteins linked with the ECM, cell proliferation, and antioxidant activity.

Conflicts of interest

The authors declare that there is no conflict of interest.

Acknowledgments

This work was financially supported by the National Research Foundation of Korea (NRF) grant funded by the Korea Government (MEST) NRF-2016K1A3A1A08953546). (NRF-2014R1A1A3050752, NRF-2015R1A2A2A01003597,

References

  1. Beissert S, Schwarz T. Mechanisms involved in ultraviolet light-induced immunosuppression. J Investig Dermatol Symp Proc 1999;4:61-4. https://doi.org/10.1038/sj.jidsp.5640183
  2. Fisher GJ, Datta SC, Talwar HS, Wang ZQ, Varani J, Kang S, Voorhees JJ. Molecular basis of sun-induced premature skin ageing and retinoid antagonism. Nature 1996;379:335-9. https://doi.org/10.1038/379335a0
  3. Jurkiewicz BA, Buettner GR. EPR detection of free radicals in UV-irradiated skin: mouse versus human. Photochem Photobiol 1996;64:918-22. https://doi.org/10.1111/j.1751-1097.1996.tb01856.x
  4. You J, Roh KB, Li Z, Liu G, Tang J, Shin S, Park D, Jung E. The antiaging properties of andrographis paniculata by activation epidermal cell stemness. Molecules 2015;20:17557-69. https://doi.org/10.3390/molecules200917557
  5. Teti A. Regulation of cellular functions by extracellular matrix. J Am Soc Nephrol 1992;2:S83-7. https://doi.org/10.1681/ASN.V210s83
  6. Di Lullo GA, Sweeney SM, Korkko J, Ala-Kokko L, San Antonio JD. Mapping the ligand-binding sites and disease-associated mutations on the most abundant protein in the human, type I collagen. J Biol Chem 2002;277:4223-31. https://doi.org/10.1074/jbc.M110709200
  7. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463-516. https://doi.org/10.1146/annurev.cellbio.17.1.463
  8. Rinnerthaler M, Bischof J, Streubel MK, Trost A, Richter K. Oxidative stress in aging human skin. Biomolecules 2015;5:545-89. https://doi.org/10.3390/biom5020545
  9. Alves Barros AS, Oliveira Carvalho H, Dos Santos IVF, Taglialegna T, Dos Santos Sampaio TI, Duarte JL, Fernandes CP, Tavares Carvalho JC. Study of the nonclinical healing activities of the extract and gel of Portulaca pilosa L. in skin wounds in wistar rats: a preliminary study. Biomed Pharmacother 2017;96:182-90. https://doi.org/10.1016/j.biopha.2017.09.142
  10. Freitas KM, Barcelos LS, Caliari MV, Salas CE, Lopes MTP. Healing activity of proteolytic fraction (P1G10) from Vasconcellea cundinamarcensis in a cutaneous wound excision model. Biomed Pharmacother 2017;96:269-78. https://doi.org/10.1016/j.biopha.2017.09.109
  11. Chen Y, Liu ZH, Xia J, Li XP, Li KQ, Xiong W, Li J, Chen DL. 20(S)-ginsenoside Rh2 inhibits the proliferation and induces the apoptosis of KG-1a cells through the $Wnt/{\beta}$eta-catenin signaling pathway. Oncol Rep 2016;36:137-46. https://doi.org/10.3892/or.2016.4774
  12. Park JY, Choi P, Kim HK, Kang KS, Ham J. Increase in apoptotic effect of Panax ginseng by microwave processing in human prostate cancer cells: in vitro and in vivo studies. J Ginseng Res 2016;40:62-7. https://doi.org/10.1016/j.jgr.2015.04.007
  13. Zhang Y, Liu QZ, Xing SP, Zhang JL. Inhibiting effect of Endostar combined with ginsenoside Rg3 on breast cancer tumor growth in tumor-bearing mice. Asian Pac J Trop Med 2016;9:180-3. https://doi.org/10.1016/j.apjtm.2016.01.010
  14. Wang CZ, Cai Y, Anderson S, Yuan CS. Ginseng metabolites on cancer chemoprevention: an angiogenesis link? Diseases 2015;3:193-204. https://doi.org/10.3390/diseases3030193
  15. Zhang H, Li Z, Zhou Z, Yang H, Zhong Z, Lou C. Antidepressant-like effects of ginsenosides: a comparison of ginsenoside Rb3 and its four deglycosylated derivatives, Rg3, Rh2, compound K, and 20(S)-protopanaxadiol in mice models of despair. Pharmacol Biochem Behav 2016;140:17-26. https://doi.org/10.1016/j.pbb.2015.10.018
  16. Lee H, Hong Y, Kwon SH, Park J, Park J. Anti-aging effects of Piper cambodianum P. Fourn. extract on normal human dermal fibroblast cells and a wound-healing model in mice. Clin Interv Aging 2016;11:1017-26. https://doi.org/10.2147/CIA.S107734
  17. Park J, Lee H, Tran Q, Mun K, Kim D, Hong Y, Kwon SH, Brazil D, Park J, Kim SH. Recognition of transmembrane protein 39A as a tumor-specific marker in brain tumor. Toxicol Res 2017;33:63-9. https://doi.org/10.5487/TR.2017.33.1.063
  18. Park J, Tran Q, Mun K, Masuda K, Kwon SH, Kim SH, Kim DH, Thomas G, Park J. Involvement of S6K1 in mitochondria function and structure in HeLa cells. Cell Signal 2016;28:1904-15. https://doi.org/10.1016/j.cellsig.2016.09.003
  19. Stephens L, Anderson K, Stokoe D, Erdjument-Bromage H, Painter GF, Holmes AB, Gaffney PR, Reese CB, McCormick F, Tempst P, et al. Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B. Science 1998;279:710-4. https://doi.org/10.1126/science.279.5351.710
  20. Huang C, Li J, Ding M, Leonard SS, Wang L, Castranova V, Vallyathan V, Shi X. UV Induces phosphorylation of protein kinase B (Akt) at Ser-473 and Thr-308 in mouse epidermal Cl 41 cells through hydrogen peroxide. J Biol Chem 2001;276:40234-40. https://doi.org/10.1074/jbc.M103684200
  21. Woodgett JR, Avruch J, Kyriakis J. The stress activated protein kinase pathway. Cancer Surv 1996;27:127-38.
  22. Briganti S, Picardo M. Antioxidant activity, lipid peroxidation and skin diseases. What's new. J Eur Acad Dermatol Venereol 2003;17:663-9. https://doi.org/10.1046/j.1468-3083.2003.00751.x
  23. Hseu YC, Lo HW, Korivi M, Tsai YC, Tang MJ, Yang HL. Dermato-protective properties of ergothioneine through induction of Nrf2/ARE-mediated antioxidant genes in UVA-irradiated Human keratinocytes. Free Radic Biol Med 2015;86:102-17. https://doi.org/10.1016/j.freeradbiomed.2015.05.026
  24. Westermarck J, Kahari VM. Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 1999;13:781-92. https://doi.org/10.1096/fasebj.13.8.781
  25. Buscher M, Rahmsdorf HJ, Litfin M, Karin M, Herrlich P. Activation of the c-fos gene by UV and phorbol ester: different signal transduction pathways converge to the same enhancer element. Oncogene 1988;3:301-11.
  26. Devary Y, Gottlieb RA, Lau LF, Karin M. Rapid and preferential activation of the c-jun gene during the mammalian UV response. Mol Cell Biol 1991;11:2804-11. https://doi.org/10.1128/MCB.11.5.2804
  27. Kock A, Schwarz T, Kirnbauer R, Urbanski A, Perry P, Ansel JC, Luger TA. Human keratinocytes are a source for tumor necrosis factor alpha: evidence for synthesis and release upon stimulation with endotoxin or ultraviolet light. J Exp Med 1990;172:1609-14. https://doi.org/10.1084/jem.172.6.1609
  28. Oxholm P, Prause JU, Schiodt M. Rational drug therapy recommendations for the treatment of patients with Sjogren's syndrome. Drugs 1998;56:345-53. https://doi.org/10.2165/00003495-199856030-00004
  29. Leverkus M, Yaar M, Eller MS, Tang EH, Gilchrest BA. Post-transcriptional regulation of UV induced TNF-alpha expression. J Invest Dermatol 1998;110:353-7. https://doi.org/10.1046/j.1523-1747.1998.00154.x
  30. Coffer PJ, Burgering BM, Peppelenbosch MP, Bos JL, Kruijer W. UV activation of receptor tyrosine kinase activity. Oncogene 1995;11:561-9.
  31. Radler-Pohl A, Sachsenmaier C, Gebel S, Auer HP, Bruder JT, Rapp U, Angel P, Rahmsdorf HJ, Herrlich P. UV-induced activation of AP-1 involves obligatory extranuclear steps including Raf-1 kinase. EMBO J 1993;12:1005-12. https://doi.org/10.1002/j.1460-2075.1993.tb05741.x
  32. Rosette C, Karin M. Ultraviolet light and osmotic stress: activation of the JNK cascade through multiple growth factor and cytokine receptors. Science 1996;274:1194-7. https://doi.org/10.1126/science.274.5290.1194
  33. Sachsenmaier C, Radler-Pohl A, Zinck R, Nordheim A, Herrlich P, Rahmsdorf HJ. Involvement of growth factor receptors in the mammalian UVC response. Cell 1994;78:963-72. https://doi.org/10.1016/0092-8674(94)90272-0
  34. Huang RP, Wu JX, Fan Y, Adamson ED. UV activates growth factor receptors via reactive oxygen intermediates. J Cell Biol 1996;133:211-20. https://doi.org/10.1083/jcb.133.1.211
  35. Xu Y, Shao Y, Zhou J, Voorhees JJ, Fisher GJ. Ultraviolet irradiation-induces epidermal growth factor receptor (EGFR) nuclear translocation in human keratinocytes. J Cell Biochem 2009;107:873-80. https://doi.org/10.1002/jcb.22195
  36. Correia M, Thiagarajan V, Coutinho I, Gajula GP, Petersen SB, Neves-Petersen MT. Modulating the structure of EGFR with UV light: new possibilities in cancer therapy. PLoS One 2014;9:e111617. https://doi.org/10.1371/journal.pone.0111617
  37. Akhlaghi M, Shabanian G, Abedinzadeh M. Special position for the anaesthetic management of a patient with giant neck and back masses. Ghana Med J 2010;44:37-8.
  38. Abedin M, King N. Diverse evolutionary paths to cell adhesion. Trends Cell Biol 2010;20:734-42. https://doi.org/10.1016/j.tcb.2010.08.002
  39. Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 2006;69:562-73. https://doi.org/10.1016/j.cardiores.2005.12.002
  40. Gordon MK, Hahn RA. Collagens. Cell Tissue Res 2010;339:247-57. https://doi.org/10.1007/s00441-009-0844-4
  41. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci 2010;123:4195-200. https://doi.org/10.1242/jcs.023820
  42. Wise SG, Weiss AS. Tropoelastin. Int J Biochem Cell Biol 2009;41:494-7. https://doi.org/10.1016/j.biocel.2008.03.017
  43. Mecham RP. Methods in elastic tissue biology: elastin isolation and purification. Methods 2008;45:32-41. https://doi.org/10.1016/j.ymeth.2008.01.007
  44. Simon MP, Pedeutour F, Sirvent N, Grosgeorge J, Minoletti F, Coindre JM, Terrier-Lacombe MJ, Mandahl N, Craver RD, Blin N, et al. Deregulation of the platelet-derived growth factor B-chain gene via fusion with collagen gene COL1A1 in dermatofibrosarcoma protuberans and giant-cell fibroblastoma. Nat Genet 1997;15:95-8. https://doi.org/10.1038/ng0197-95
  45. Zaidi SK, Young DW, Montecino M, Lian JB, Stein JL, van Wijnen AJ, Stein GS. Architectural epigenetics: mitotic retention of mammalian transcriptional regulatory information. Mol Cell Biol 2010;30:4758-66. https://doi.org/10.1128/MCB.00646-10
  46. Yuan W, Yufit T, Li L, Mori Y, Chen SJ, Varga J. Negative modulation of alpha1(I) procollagen gene expression in human skin fibroblasts: transcriptional inhibition by interferon-gamma. J Cell Physiol 1999;179:97-108. https://doi.org/10.1002/(SICI)1097-4652(199904)179:1<97::AID-JCP12>3.0.CO;2-E
  47. Bhogal RK, Bona CA. Regulatory effect of extracellular signal-regulated kinases (ERK) on type I collagen synthesis in human dermal fibroblasts stimulated by IL-4 and IL-13. Int Rev Immunol 2008;27:472-96. https://doi.org/10.1080/08830180802430974
  48. Hanson KM, Clegg RM. Observation and quantification of ultraviolet-induced reactive oxygen species in ex vivo human skin. Photochem Photobiol 2002;76:57-63. https://doi.org/10.1562/0031-8655(2002)0760057OAQOUI2.0.CO2
  49. Heck DE, Vetrano AM, Mariano TM, Laskin JD. UVB light stimulates production of reactive oxygen species: unexpected role for catalase. J Biol Chem 2003;278:22432-6. https://doi.org/10.1074/jbc.C300048200
  50. Sakurai H, Yasui H, Yamada Y, Nishimura H, Shigemoto M. Detection of reactive oxygen species in the skin of live mice and rats exposed to UVA light: a research review on chemiluminescence and trials for UVA protection. Photochem Photobiol Sci 2005;4:715-20. https://doi.org/10.1039/b417319h
  51. Fernandez-Garcia E. Skin protection against UV light by dietary antioxidants. Food Funct 2014;5:1994-2003. https://doi.org/10.1039/C4FO00280F
  52. Barata C, Varo I, Navarro JC, Arun S, Porte C. Antioxidant enzyme activities and lipid peroxidation in the freshwater cladoceran Daphnia magna exposed to redox cycling compounds. Comp Biochem Physiol C Toxicol Pharmacol 2005;140:175-86. https://doi.org/10.1016/j.cca.2005.01.013
  53. Coyle CH, Martinez LJ, Coleman MC, Spitz DR, Weintraub NL, Kader KN. Mechanisms of H2O2-induced oxidative stress in endothelial cells. Free Radic Biol Med 2006;40:2206-13. https://doi.org/10.1016/j.freeradbiomed.2006.02.017
  54. Min JK, Kim JH, Cho YL, Maeng YS, Lee SJ, Pyun BJ, Kim YM, Park JH, Kwon YG. 20(S)-Ginsenoside Rg3 prevents endothelial cell apoptosis via inhibition of a mitochondrial caspase pathway. Biochem Biophys Res Commun 2006;349:987-94. https://doi.org/10.1016/j.bbrc.2006.08.129
  55. Purdom-Dickinson SE, Lin Y, Dedek M, Morrissy S, Johnson J, Chen QM. Induction of antioxidant and detoxification response by oxidants in cardiomyocytes: evidence from gene expression profiling and activation of Nrf2 transcription factor. J Mol Cell Cardiol 2007;42:159-76. https://doi.org/10.1016/j.yjmcc.2006.09.012
  56. Zhu H, Itoh K, Yamamoto M, Zweier JL, Li Y. Role of Nrf2 signaling in regulation of antioxidants and phase 2 enzymes in cardiac fibroblasts: protection against reactive oxygen and nitrogen species-induced cell injury. FEBS Lett 2005;579:3029-36. https://doi.org/10.1016/j.febslet.2005.04.058
  57. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 2009;284:13291-5. https://doi.org/10.1074/jbc.R900010200
  58. Li W, Khor TO, Xu C, Shen G, Jeong WS, Yu S, Kong AN. Activation of Nrf2-antioxidant signaling attenuates NFkappaB-inflammatory response and elicits apoptosis. Biochem Pharmacol 2008;76:1485-9. https://doi.org/10.1016/j.bcp.2008.07.017
  59. Liu C, Vojnovic D, Kochevar IE, Jurkunas UV. UV-A irradiation activates Nrf2-regulated antioxidant defense and induces p53/caspase3-dependent apoptosis in corneal endothelial cells. Invest Ophthalmol Vis Sci 2016;57:2319-27. https://doi.org/10.1167/iovs.16-19097
  60. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013;53:401-26. https://doi.org/10.1146/annurev-pharmtox-011112-140320
  61. Zhong JL, Edwards GP, Raval C, Li H, Tyrrell RM. The role of Nrf2 in ultraviolet A mediated heme oxygenase 1 induction in human skin fibroblasts. Photochem Photobiol Sci 2010;9:18-24. https://doi.org/10.1039/B9PP00068B
  62. Okamoto K, Shaw JM. Mitochondrial morphology and dynamics in yeast and multicellular eukaryotes. Annu Rev Genet 2005;39:503-36. https://doi.org/10.1146/annurev.genet.38.072902.093019
  63. Hoppins S, Lackner L, Nunnari J. The machines that divide and fuse mitochondria. Annu Rev Biochem 2007;76:751-80. https://doi.org/10.1146/annurev.biochem.76.071905.090048
  64. Westermann B. Molecular machinery of mitochondrial fusion and fission. J Biol Chem 2008;283:13501-5. https://doi.org/10.1074/jbc.R800011200
  65. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E. Mitochondrial fragmentation in neurodegeneration. Nat Rev Neurosci 2008;9:505-18. https://doi.org/10.1038/nrn2417
  66. Galluzzi L, Morselli E, Kepp O, Kroemer G. Targeting post-mitochondrial effectors of apoptosis for neuroprotection. Biochim Biophys Acta 2009;1787:402-13. https://doi.org/10.1016/j.bbabio.2008.09.006
  67. Tait LF, Ortega G, Tran DD, Fullum TM. Resolution of Uncontrolled Type 2 Diabetes after Laparoscopic Truncal Vagotomy, Subtotal Gastrectomy, and Roux-en-Y Gastrojejunostomy for a Patient with Intractable Gastric Ulcers. Case Rep Surg 2012;2012:102752. https://doi.org/10.1155/2012/102752
  68. Gniadecki R, Thorn T, Vicanova J, Petersen A, Wulf HC. Role of mitochondria in ultraviolet-induced oxidative stress. J Cell Biochem 2000;80:216-22. https://doi.org/10.1002/1097-4644(20010201)80:2<216::AID-JCB100>3.0.CO;2-H
  69. Birch-Machin MA, Swalwell H. How mitochondria record the effects of UV exposure and oxidative stress using human skin as a model tissue. Mutagenesis 2010;25:101-7. https://doi.org/10.1093/mutage/gep061
  70. Bowman A, Birch-Machin MA. Mitochondrial DNA as a biosensor of UV exposure in human skin. Methods Mol Biol 2015;1265:379-88. https://doi.org/10.1007/978-1-4939-2288-8_27
  71. Skulachev VP. Mitochondrial filaments and clusters as intracellular powertransmitting cables. Trends Biochem Sci 2001;26:23-9. https://doi.org/10.1016/S0968-0004(00)01735-7
  72. Frank S. Dysregulation of mitochondrial fusion and fission: an emerging concept in neurodegeneration. Acta Neuropathol 2006;111:93-100. https://doi.org/10.1007/s00401-005-0002-3
  73. Abdelwahid E, Pelliniemi LJ, Jokinen E. Cell death and differentiation in the development of the endocardial cushion of the embryonic heart. Microsc Res Tech 2002;58:395-403. https://doi.org/10.1002/jemt.10159
  74. Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal 2009;3:287-310. https://doi.org/10.1007/s12079-009-0075-1

Cited by

  1. Red Ginseng Attenuates Aβ-Induced Mitochondrial Dysfunction and Aβ-mediated Pathology in an Animal Model of Alzheimer’s Disease vol.20, pp.12, 2019, https://doi.org/10.3390/ijms20123030
  2. Relationship Between Ginsenoside Rg3 and Metabolic Syndrome vol.11, 2019, https://doi.org/10.3389/fphar.2020.00130
  3. Herbal Medicine for Cardiovascular Diseases: Efficacy, Mechanisms, and Safety vol.11, 2019, https://doi.org/10.3389/fphar.2020.00422
  4. Terpenoids as Potential Geroprotectors vol.9, pp.6, 2019, https://doi.org/10.3390/antiox9060529
  5. Ca 2+ -activated mitochondrial biogenesis and functions improve stem cell fate in Rg3-treated human mesenchymal stem cells vol.11, pp.1, 2020, https://doi.org/10.1186/s13287-020-01974-3
  6. 인삼 뷰티 모노그래프: 인삼의 역사와 피부 효능 연구에 관한 고찰 vol.10, pp.12, 2019, https://doi.org/10.22156/cs4smb.2020.10.12.166
  7. Antioxidative and Skin Protective Effects of Canarium subulatum Methanol Extract on Keratinocytes vol.2021, 2021, https://doi.org/10.1155/2021/6692838
  8. Panax ginseng C. A. Meyer Phenolic Acid Extract Alleviates Ultraviolet B-Irradiation-Induced Photoaging in a Hairless Mouse Skin Photodamage Model vol.2021, 2019, https://doi.org/10.1155/2021/9962007
  9. Review of ginsenosides targeting mitochondrial function to treat multiple disorders: Current status and perspectives vol.45, pp.3, 2021, https://doi.org/10.1016/j.jgr.2020.12.004
  10. Korean Red Ginseng attenuates ultraviolet-mediated inflammasome activation in keratinocytes vol.45, pp.3, 2021, https://doi.org/10.1016/j.jgr.2021.02.002
  11. Ginsenoside Rg3 Alleviates Aluminum Chloride-Induced Bone Impairment in Rats by Activating the TGF-β1/Smad Signaling Pathway vol.69, pp.43, 2021, https://doi.org/10.1021/acs.jafc.1c04695
  12. Protective effects of galangin against H2O2/UVB-induced dermal fibroblast collagen degradation via hsa-microRNA-4535-mediated TGFβ/Smad signaling vol.13, pp.23, 2019, https://doi.org/10.18632/aging.203750