Open Access

Dracorhodin perchlorate enhances wound healing via β‑catenin, ERK/p38, and AKT signaling in human HaCaT keratinocytes

  • Authors:
    • Chi-Cheng Lu
    • Jai-Sing Yang
    • Yu-Jen Chiu
    • Fuu-Jen Tsai
    • Yuan-Man Hsu
    • Mei-Chin Yin
    • Yu-Ning Juan
    • Tsung-Jung Ho
    • Hao-Ping Chen
  • View Affiliations

  • Published online on: June 2, 2021     https://doi.org/10.3892/etm.2021.10254
  • Article Number: 822
  • Copyright: © Lu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Dracorhodin can be isolated from the exudates of the fruit of Daemonorops draco. Previous studies suggested that dracorhodin perchlorate can promote fibroblast proliferation and enhance angiogenesis during wound healing. In the present study, the potential bioactivity of dracorhodin perchlorate in human HaCaT keratinocytes, were investigated in vitro, with specific focus on HaCaT wound healing. The results of in vitro scratch assay demonstrated the progressive closure of the wound after treatment with dracorhodin perchlorate in a time‑dependent manner. An MTT assay and propidium iodide exclusion detected using flow cytometry were used to detect cell viability of HaCaT cells. Potential signaling pathways underlying the effects mediated by dracorhodin perchlorate in HaCaT cells were clarified by western blot analysis and kinase activity assays. Dracorhodin perchlorate significantly increased the protein expression levels of β‑catenin and activation of AKT, ERK and p38 in HaCaT cells. In addition, dracorhodin perchlorate did not induce HaCaT cell proliferation but promoted cell migration. Other mechanisms may yet be involved in the dracorhodin perchlorate‑induced wound healing process of human keratinocytes. In summary, dracorhodin perchlorate may serve to be a potential molecularly‑targeted phytochemical that can improve skin wound healing.

Introduction

Dragon blood is a type of deep-red resin that has been used as a form of traditional medicine worldwide since ancient times (1). Families of plants with the ability to produce dragon blood can be classified into four different categories: Asparagaceae, Arecaceae, Chamaesyce and Fabaceae (Table I) (2-4). The earliest record of dragon blood being used for medicine was performed using the plant genus Dracaena in Socotra Island in Yemen (5). Due to being situated in ideal location and the rapid growth rate of the palm tree species Daemonorops draco, dragon blood extracted from this tree became the mainstream product in the traditional Chinese medicine (TCM) market after the Ming Dynasty in China (6). Therefore, red resin from Daemonorops draco is recognized as authentic dragon blood in the official documents of China, Hong Kong and Taiwan (7-9). In particular, dracorhodin has become the reference standard of dragon blood from the palm tree D. draco (10), where the content of dracorhodin in dragon blood for medical use is recommended to be >1% (9).

Table I

Plant species that contain dragon blood and their respective geographical origins (4).

Table I

Plant species that contain dragon blood and their respective geographical origins (4).

Plant familyPlant speciesOrigin
AsparagaceaeDracaena cinnabariSocotra Island, Yemen
 Dracaena dracoCanary Islands, Spain
 Dracaena cochinchinensisYunnan Province, China
ArecaceaeDaemonorops dracoIndonesia and Malaysia
ChamaesyceCroton salutaris 
 C. lechleriAmazon basin
 C. draconoides 
FabaceaePterocarpus dracoIndia
 P. marsupium 

Dragon blood has been included in recipes for wound treatment in various ancient traditional Chinese medical guides of the 14-18th century (6). Accumulating evidence has also indicated that dragon blood possesses wound healing activities both in vivo and in vitro (1,11-13). Accordingly, a previous study showed that the herbal complex ‘Jinchuang Ointment’ can successfully treat non-healing wounds on patients with diabetes, where dragon blood from D. draco is one of the components (2.1%) in this ointment (14).

Dracorhodin perchlorate is a synthetic chemical analog of dracorhodin (Fig. 1) (10,15-18) that has been shown to exhibit angiogenic activity on human umbilical vein endothelial cells (10,19). Recently, both dracorhodin perchlorate and methanol extracts of crude dragon blood have demonstrated in vivo pro-angiogenic activity in zebrafish embryos (10). Additionally, previous studies have revealed bioactivities of dracorhodin perchlorate in human cancer cells, where it can induce apoptotic cell death in different types of cancer cells, including U87MG and T98G glioma cells (20), SK-MES-1 human lung squamous carcinoma cells (21), MCF-7 human breast cancer cells (22), SGC-7901 human gastric tumor cells (23), PC-3 human prostate cancer cells (18), HL-60 leukemia cells (16), A375-S2 human melanoma cells (24) and HeLa human cervical cancer cells (15,25). Furthermore, dracorhodin perchlorate was reported to promote NIH-3T3 fibroblast cell proliferation in vitro and induce rat wound healing in vivo (26). Dracorhodin perchlorate has also been shown to accelerate skin wound healing in Wistar rats in vivo (27). However, the effects of dracorhodin perchlorate on wound healing in human HaCaT keratinocytes remain poorly understood. Therefore, in the present study the underlying mechanism of the wounding healing regulation and associated signaling pathways mediated by dracorhodin perchlorate in HaCaT keratinocytes was investigated in vitro.

Materials and methods

Reagents and chemicals

DMEM, FBS, penicillin/streptomycin and L-glutamine were purchased from HyClone, Cytiva. All primary antibodies and anti-mouse/-rabbit immunoglobulin IgG HRP-linked secondary antibodies were procured from GeneTex International Corporation. Reagent grade DMSO, thiazolyl blue tetrazolium bromide (MTT), PBS, propidium iodide (PI), wortmannin (an AKT inhibitor), U0126 (an ERK inhibitor) and SB203580 (a p38 inhibitor), were obtained from Sigma-Aldrich, Merck KGaA unless otherwise specified. Dracorhodin perchlorate (batch no. 110811-201506; purity, 98.6%) was purchased from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China), which was dissolved in DMSO and stored at -20˚C freezer before use. The vehicle control was kept below 0.1% DMSO in culture medium.

Cell culture

Human skin HaCaT keratinocytes were obtained from CLS Cell Lines Service GmbH and cultured in DMEM supplemented with 10% FBS, 100 U/ml penicillin, 100 µg/ml streptomycin and 2 mM L-glutamine in a humidified atmosphere at 37˚C in 5% CO2/95% air as previously described (28,29). HaCaT cells were cultured until passage 40 (20 weeks) and did not exceed 40 generations.

Dynamic cell migration assay

HaCaT cells (1x104 cells/well) were seeded into a 96-well plate overnight before the wound area (700-800-µm wide) was created using Incucyte 96-Well Woundmaker Tool (cat. no. 4563; Essen BioScience). The scratched cells were then exposed to 0, 1 and 2 µg/ml dracorhodin perchlorate in serum-free DMEM (HyClone; Cytiva). The cell migration experiment was conducted over 24 h with data collection every 3 h at 37˚C. The cell images and wound width were monitored using the IncuCyte ZOOM System instrument by light microscopy (magnification, x100) [(Incucyte S3 Live-Cell Analysis System and Incucyte Scratch Wound Analysis Software Module (cat. no. 9600-0012; Essen BioScience)] as previously described (30,31).

Cell viability assay

HaCaT cells (2.5x105 cells/well) were plated into 24-well plates and treated with or without 0.5, 1 and 2 µg/ml dracorhodin perchlorate for 24 h at 37˚C. Cell viability was determined using MTT assay as previously described (32-34). In brief, MTT solution (0.5 mg/ml) was added to each well for 2 h at 37˚C before the blue formazan crystals were dissolved in DMSO (500 µl/well) by constant shaking for 10 min. The absorbance of each well was measured using an ELISA plate reader at a test wavelength of 570 nm with a reference wavelength of 620 nm.

For the PI exclusion assay, 2.5x105 cells/well were harvested and plated into 24-well plates and resuspended in 500 µl of 5 µg/ml PI for 5 min at 4˚C. The numbers of viable cells and dead cells were determined using a BD FACSCalibur™ Flow Cytometry System and BD CellQuest Pro Software version 6.0 (both BD Biosciences), where the PI dye solution was applied to specifically stain dead cells. Cell viability was calculated as follows: % Cell viability = (PIem of test)/(PIem of control) x100%. Where, PIem = gated cells of PI fluorescence emission (<102 fluorescence).

Western blot analysis

HaCaT cells (5x106 cells per 75T flask) were incubated with or without 1 and 2 µg/ml dracorhodin perchlorate for 12 h at 37˚C. Cell samples were lysed in Trident RIPA Lysis Buffer (GeneTex International Corporation) and collected as previously described (35,36). Protein concentration was determined using the Pierce bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc.). Equal amounts of the protein sample (40 µg) were prepared, and 10-12% SDS-PAGE was performed. Proteins were transferred onto an Immobilon-P polyvinylidene difluoride transfer membrane prior to blocking with Trident Universal Protein Blocking Reagent (GeneTex International Corporation) for 1 h at room temperature. The membrane was subsequently incubated with primary antibodies against β-catenin (cat. no. GTX101435), phosphorylated (p)-AKT (Ser473) (cat. no. GTX28932), AKT (cat. no. GTX121937), p-ERK (cat. no. GTX59568), ERK (cat. no. GTX59618), p38 (cat. no. GTX110720), p-p38 (cat. no. GTX48614) and β-actin (cat. no. GTX109639) at a dilution of 1:1,000 at 4˚C overnight. The membranes were then incubated with the appropriate anti-mouse (cat. no. GTX213111-01) and anti-rabbit (cat. no. GTX213110-01) IgG HRP-linked secondary antibodies at a dilution of 1:10,000 for 1 h at room temperature. Blot visualization was performed using the Immobilon Western Chemiluminescent HRP Substrate (Merck KGaA) and all bands of immunoblots were normalized to the densitometric value of β-actin. These experiments were conducted in duplicate. The bands were quantified by densitometry using ImageJ software version 1.41 (National Institutes of Health) as previously described (33,37-39).

Sandwich ELISA assay for p-protein kinases

HaCaT cells (5x106 cells per T75 flask) were treated with or without 0.5, 1 and 2 µg/ml dracorhodin perchlorate for 12 h at 37˚C. The cells were harvested and total proteins were collected using Trident RIPA Lysis Buffer (GeneTex International Corporation). The samples were incubated in microwells coated with the appropriate antibody for 2 h at 37˚C (for p-ERK) or overnight at 4˚C (for p-Akt) according to the manufacturer's protocols of the PathScan Sandwich ELISA kits [p-Akt (Thr308; cat. no. 7252C) and p-ERK (Thr202/Tyr204; cat. no. 7177C); Cell Signaling Technology, Inc.]. The secondary antibodies and the reagents used for the chemical reactions were provided in these kits. Absorbance was measured using an ELISA reader (Anthos 2001; Anthos Labtec Instruments GmbH) at a wavelength of 450 nm, as previously described (38-40).

Wound healing assay

HaCaT cells were placed in a six-well tissue culture plates for 24 h at 37˚C and cultured to 90% confluence. Individual wells were then scratched with a 200-µl micropipette tip to create a denuded zone of constant width (1 mm). Cells were then cultured in serum-free DMEM (HyClone; Cytiva) and incubated with or without dracorhodin perchlorate (1 µg/ml) or with specific protein kinase inhibitors [10 µM wortmannin (AKT inhibitor), 10 µM U0126 (ERK inhibitor) or 10 µM SB203580 (p38 inhibitor)] and 0.1% DMSO as a control for 24 h at 37˚C. To determine cell migration, cell images were captured after 24 h treatment under phase-contrast microscopy (magnification, x100; Leica DMIL microscope; type 090-135.001; Leica Microsystems GmbH), as previously described (38). Image analysis was performed by using AxioVision LE64 software (version 4.9.1.0; Carl Zeiss AG). The ability of cell migration was calculated as follows: Cell migration (% of control) = [Gw (0 h) of test-Gw (24 h) of test]/[Gw (0 h) of control-Gw (24 h) of control] x100%. Where, Gw (0 h) = gap width at 0 h, and Gw (24 h) = gap width at 24 h.

Statistical analysis

All results are expressed as the mean ± standard deviation (three experimental repeats per assay). Differences among groups were determined by one-way analysis of variance followed by Tukey's or Dunnett's post hoc test with SPSS software version 16.0 (SPSS, Inc.). P<0.05 or P<0.001 was considered to indicate a statistically significant difference.

Results

Dracorhodin perchlorate promotes cell migration in HaCaT keratinocytes

Wound healing activity was first investigated, which mimics the migration of keratinocytes toward wound margins in vitro. As shown in Fig. 2A, the wound was closing progressively following treatment with dracorhodin perchlorate in HaCaT cells. No significant difference was found between 1 and 2 µg/ml groups. Therefore, the optimal dracorhodin perchlorate concentration to monitor wound healing activity was found to be 1 µg/ml (Fig. 2B). Quantitative data indicated that dracorhodin perchlorate at 1 µg/ml significantly reduced the wound width at the time point of 24 h and markedly promoted HaCaT cell migration compared with those in control cells (Fig. 2C). Therefore, this suggests that dracorhodin perchlorate enhanced cell migration in HaCaT keratinocytes.

Dracorhodin perchlorate does not enhance cell viability in HaCaT keratinocytes

As previously reported, the cell viability of human primary fibroblast proliferation significantly increased by dracorhodin perchlorate treatment (41). Therefore, MTT assay and PI exclusion method were performed to investigate the cell viability and cytotoxicity of HaCaT keratinocytes after dracorhodin perchlorate exposure. Dracorhodin perchlorate did not affect the viability of HaCaT cells at the highest concentration tested in this study, which was 2 µg/ml (Fig. 3A). As shown in Fig. 3B and C, viable cells were generally excluded from PI because of low emission fluorescence. No significant change was observed in dracorhodin perchlorate-treated HaCaT keratinocytes (Fig. 3B). Moreover, flow cytometry results showed that the total counts of dead and viable cells were <10 and >90%, respectively (Fig. 3C). Thus, the migration ability of HaCaT cells caused by dracorhodin perchlorate was not due to the cell viability of HaCaT keratinocytes.

Dracorhodin perchlorate increases the protein expression of β-catenin and phosphorylation of Akt, p38 and ERK in HaCaT keratinocytes

Previous studies have suggested that wound healing is closely associated with β-catenin signaling (42,43), where keratinocyte mobility can be stimulated through the MAPK and AKT signaling pathways (44-46). Western blotting experiments were therefore performed to investigate the effects of dracorhodin perchlorate treatment on the protein expression of β-catenin and both phosphorylation of ERK and AKT. Dracorhodin perchlorate treatment (2 µg/ml for 12 h) markedly upregulated the protein levels of β-catenin, phosphorylated ERK, phosphorylated Akt and phosphorylated p38 in HaCaT keratinocytes compared with those in control cells (Fig. 4). This was subsequently verified using sandwich ELISA. The levels of both phosphorylated AKT and ERK significantly increased after dracorhodin perchlorate treatment in a dose-dependent manner (Fig. 5).

To confirm further that the mechanism of wound healing underlying the effects of dracorhodin perchlorate, wortmannin, U0126 and SB203580, inhibitors of AKT, ERK and p38 MAPK, respectively, were used. As shown in Fig. 6, inhibitors of AKT, ERK and p38 MAPK could all significantly inhibit the migration of dracorhodin perchlorate-treated keratocytes. Therefore, it could be concluded that dracorhodin perchlorate activated wound healing by upregulating the ERK, MAPK and AKT signaling pathways in human HaCaT keratinocytes in vitro.

Discussion

Dracorhodin perchlorate is a new synthetic chemical analog of dracorhodin (10,15-18). Dracorhodin is originally the fruit extract of dragon blood, which can be used as a TCM (2,10). Dragon blood confers a number of bioactivities, including antibacterial, anti-diarrheal, anti-inflammatory, anti-oxidant, antitumor, anti-ulcer, antiviral, immune-modulatory and wound healing functions (1). Previous studies have demonstrated that dragon blood can promote the proliferation of fibroblasts, increase the ratio of cells in the S-phase of the cell cycle and collagen formation, in addition to enhancing wound healing (1,11,12). In the present study, dracorhodin perchlorate was found to significantly promote wound healing in human HaCaT keratinocytes. However treatment with dracorhodin perchlorate (0.5-2 µg/ml) in HaCaT keratinocytes exerted no statistically significant effects on keratinocyte viability. To the best of our knowledge, the present study is the first time an in vitro scratch assay was utilized using dynamic analysis facilitated by the IncuCyte Kinetic Live Cell Imaging System to evaluate the effects on wound closure in dracorhodin perchlorate-treated HaCaT cells. Furthermore, visible gaps were observed in the monolayer of cells at the start of the assay. In accordance with this result, the cell number did not increase.

Previous studies have shown that activation of the Wnt/β-catenin signaling pathway serves an important role in the wound healing process of human keratinocytes (47). During the wound healing of keratinocytes, cell migration and invasion are regulated by the activation of the PI3K/AKT, ERK, p38/MAPK and Wnt/β-catenin signaling cascades (48-51). Additionally, activation of the ERK and p38/MAPK pathways in cell migration is crucial for wound healing (52,53). In accordance with these previous findings aforementioned, results from the present study showed that dracorhodin perchlorate treatment markedly increased the phosphorylation of AKT, ERK and p38 and kinase activities in HaCaT cells. In addition, dracorhodin perchlorate-treated HaCaT cells were selectively incubated with the AKT inhibitor wortmannin, the ERK inhibitor U0126 or the p38 inhibitor SB203580 for 24 h. Cell migration was found to be inhibited by all three inhibitors, suggesting that the ERK, p38 MAPK and AKT signaling pathways were all involved. All these results indicated that AKT, ERK and p38 MAPK were activated by dracorhodin perchlorate treatment during the wound healing process in HaCaT cells in a dose-dependent manner within the concentration range of 0 and 2 µg/ml. However, the optimal dracorhodin perchlorate concentration for wound healing activity was found in the present study to be 1 µg/ml rather than 2 µg/ml. Therefore, other mechanisms may be involved in this dracorhodin perchlorate-induced wound healing process in HaCaT cells.

A proposed summary of the wound healing mechanism underlying the effects mediated by dracorhodin perchlorate in HaCaT cells is shown in Fig. 7. It was found that dracorhodin perchlorate promoted the wound healing activity of HaCaT keratinocytes through the β-catenin, ERK, p38 MAPK and AKT signaling pathways. These findings provide novel insights into the mechanistic information in the wound healing activity of dracorhodin perchlorate in human HaCaT keratinocytes.

Acknowledgements

The authors would like to acknowledge the work of Mr. Meng-Jou Liao and Mr. Chin-Chen Lin (Tekon Scientific Corporation, Taipei, Taiwan) for their assistance and equipment support on this study. The authors would also like to thank Mr. Chang-Wei Li (AllBio Science, Inc., Taichung, Taiwan) for his excellent technical support.

Funding

Funding: Financial support was provided by the China Medical University Hospital (grant no. DMR-106-179) and the China Medical University Beigang Hospital (grant no. CMUBH R106-005).

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

Conceptualization and study design: TJH and HPC. Cell migration, cell viability, western blotting and protein kinase activity: CCL, JSY and YJC. Protein kinase inhibition: CCL and YNJ. Wound healing assay and acquisition of data: YMH, MCY and FJT. Statistical analysis of all data and interpretation of results: FJT, TJH and HPC. CCL and HPC confirm the authenticity of all the raw data. 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 

Pona A, Cline A, Kolli SS, Taylor SL and Feldman SR: Review of future insights of Dragon's Blood in dermatology. Dermatol Ther. 32(e12786)2019.PubMed/NCBI View Article : Google Scholar

2 

Sousa MM, Melo MJ, Parola AJ, Seixas de Melo JS, Catarino F, Pina F, Cook FE, Simmonds MS and Lopes JA: Flavylium chromophores as species markers for dragon's blood resins from Dracaena and Daemonorops trees. J Chromatogr A. 1209:153–161. 2008.PubMed/NCBI View Article : Google Scholar

3 

Edwards HG, de Oliveira LF and Prendergast HD: Raman spectroscopic analysis of dragon's blood resins-basis for distinguishing between Dracaena (Convallariaceae), Daemonorops (Palmae) and Croton (Euphorbiaceae). Analyst. 129:134–138. 2004.PubMed/NCBI View Article : Google Scholar

4 

Wu C, Cai XQ, Chang Y, Chen CH, Ho TJ, Lai SC and Chen HP: Rapid identification of dragon blood samples from Daemonorops draco, Dracaena cinnabari and Dracaena cochinchinensis by MALDI-TOF mass spectrometry. Phytochem Anal. 30:720–726. 2019.PubMed/NCBI View Article : Google Scholar

5 

Al-Okaishi A: Exploring the historical distribution of Dracaena cinnabari using ethnobotanical knowledge on Socotra Island, Yemen. J Ethnobiol Ethnomed. 17(22)2021.PubMed/NCBI View Article : Google Scholar

6 

Wu KC: Quality evaluation of herbal medicine Ercha, Xuejie, Ruxiang and Moyao. Master Thesis, China Medical University, 2011.

7 

Department of Health, Hong Kong Special Administrative Region: Hong Kong Chinese Materia Medica strandards. The People's Republic of China: Department of Health, Hong Kong Special Administrative Region, 362-368, 2005.

8 

State Pharmacopoeia Commission of the PRC: Pharmacopoeia of the People's Republic of China. 1 Beijing, People's Medical Publishing House, p142, 2015.

9 

Taiwan Herbal Pharmacopeia Editorial Panel Committee. Taiwan Herbal Pharmacopeia, version 2. Taipei: Ministry of Health and Welfare, Executive Yuan, pp102-103, 2013.

10 

Krishnaraj P, Chang Y, Ho TJ, Lu NC, Lin MD and Chen HP: In vivo pro-angiogenic effects of dracorhodin perchlorate in zebrafish embryos: A novel bioactivity evaluation platform for commercial dragon blood samples. J Food Drug Anal. 27:259–265. 2019.PubMed/NCBI View Article : Google Scholar

11 

Namjoyan F, Kiashi F, Moosavi ZB, Saffari F and Makhmalzadeh BS: Efficacy of Dragon's blood cream on wound healing: A randomized, double-blind, placebo-controlled clinical trial. J Tradit Complement Med. 6:37–40. 2016.PubMed/NCBI View Article : Google Scholar

12 

Li D, Hui R, Hu Y, Han Y and Guo S: Effects of extracts of Dragon's blood on fibroblast proliferation and extracellular matrix hyaluronic acid. Zhonghua Zheng Xing Wai Ke Za Zhi. 31:53–57. 2015.PubMed/NCBI(In Chinese).

13 

Xin N, Yang FJ, Li Y, Li YJ, Dai RJ, Meng WW, Chen Y and Deng YL: Dragon's blood dropping pills have protective effects on focal cerebral ischemia Rats model. Phytomedicine. 21:68–74. 2013.PubMed/NCBI View Article : Google Scholar

14 

Ho TJ, Jiang SJ, Lin GH, Li TS, Yiin LM, Yang JS, Hsieh MC, Wu CC, Lin JG and Chen HP: The in vitro and in vivo Wound Healing Properties of the Chinese Herbal medicine ‘Jinchuang Ointment’. Evid Based Complement Alternat Med. 2016(1654056)2016.PubMed/NCBI View Article : Google Scholar

15 

Xia M, Wang D, Wang M, Tashiro S, Onodera S, Minami M and Ikejima T: Dracorhodin perchlorate induces apoptosis via activation of caspases and generation of reactive oxygen species. J Pharmacol Sci. 95:273–283. 2004.PubMed/NCBI View Article : Google Scholar

16 

Xia MY, Wang MW, Cui Z, Tashiro SI, Onodera S, Minami M and Ikejima T: Dracorhodin perchlorate induces apoptosis in HL-60 cells. J Asian Nat Prod Res. 8:335–343. 2006.PubMed/NCBI View Article : Google Scholar

17 

Wang Y, Wang Q, Liu J, Zhang L and Zhang B: Dracorhodin perchlorate inhibit high glucose-induced connective tissue growth factor formation in human mesangial cells. Zhongguo Zhong Yao Za Zhi. 34:896–899. 2009.PubMed/NCBI(In Chinese).

18 

He Y, Ju W, Hao H, Liu Q, Lv L and Zeng F: Dracorhodin perchlorate suppresses proliferation and induces apoptosis in human prostate cancer cell line PC-3. J Huazhong Univ Sci Technolog Med Sci. 31(215)2011.PubMed/NCBI View Article : Google Scholar

19 

Li F, Jiang T, Liu W, Hu Q and Yin H: The angiogenic effect of dracorhodin perchlorate on human umbilical vein endothelial cells and its potential mechanism of action. Mol Med Rep. 14:1667–1672. 2016.PubMed/NCBI View Article : Google Scholar

20 

Chen X, Luo J, Meng L, Pan T, Zhao B, Tang ZG and Dai Y: Dracorhodin perchlorate induces the apoptosis of glioma cells. Oncol Rep. 35:2364–2372. 2016.PubMed/NCBI View Article : Google Scholar

21 

Zhang G, Sun M, Zhang Y, Hua P, Li X, Cui R and Zhang X: Dracorhodin perchlorate induces G1/G0 phase arrest and mitochondria-mediated apoptosis in SK-MES-1 human lung squamous carcinoma cells. Oncol Lett. 10:240–246. 2015.PubMed/NCBI View Article : Google Scholar

22 

Yu JH, Zheng GB, Liu CY, Zhang LY, Gao HM, Zhang YH, Dai CY, Huang L, Meng XY, Zhang WY and Yu XF: Dracorhodin perchlorate induced human breast cancer MCF-7 apoptosis through mitochondrial pathways. Int J Med Sci. 10:1149–1156. 2013.PubMed/NCBI View Article : Google Scholar

23 

Rasul A, Ding C, Li X, Khan M, Yi F, Ali M and Ma T: Dracorhodin perchlorate inhibits PI3K/Akt and NF-κB activation, up-regulates the expression of p53, and enhances apoptosis. Apoptosis. 17:1104–1119. 2012.PubMed/NCBI View Article : Google Scholar

24 

Xia M, Wang M, Tashiro S, Onodera S, Minami M and Ikejima T: Dracorhodin perchlorate induces A375-S2 cell apoptosis via accumulation of p53 and activation of caspases. Biol Pharm Bull. 28:226–232. 2005.PubMed/NCBI View Article : Google Scholar

25 

Xia MY, Wang MW, Wang HR, Tashiro S and Ikejima T: Mechanism of dracorhodin perchlorate-induced Hela cell apoptosis. Yao Xue Xue Bao. 39:966–970. 2004.PubMed/NCBI(In Chinese).

26 

Jiang X, Liu L, Qiao L, Zhang B, Wang X, Han Y and Yu W: Dracorhodin perchlorate regulates fibroblast proliferation to promote rat's wound healing. J Pharmacol Sci. 136:66–72. 2018.PubMed/NCBI View Article : Google Scholar

27 

Jiang XW, Qiao L, Liu L, Zhang BQ, Wang XW, Han YW and Yu WH: Dracorhodin Perchlorate Accelerates Cutaneous Wound Healing in Wistar Rats. Evid Based Complement Alternat Med. 2017(8950516)2017.PubMed/NCBI View Article : Google Scholar

28 

Boukamp P, Petrussevska RT, Breitkreutz D, Hornung J, Markham A and Fusenig NE: Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell line. J Cell Biol. 106:761–771. 1988.PubMed/NCBI View Article : Google Scholar

29 

Xia Q, Chiang HM, Yin JJ, Chen S, Cai L, Yu H and Fu PP: UVA photoirradiation of benzo[a]pyrene metabolites: Induction of cytotoxicity, reactive oxygen species, and lipid peroxidation. Toxicol Ind Health. 31:898–910. 2015.PubMed/NCBI View Article : Google Scholar

30 

Gelles JD and Chipuk JE: Robust high-throughput kinetic analysis of apoptosis with real-time high-content live-cell imaging. Cell Death Dis. 7(e2493)2016.PubMed/NCBI View Article : Google Scholar

31 

Lee MR, Lin C, Lu CC, Kuo SC, Tsao JW, Juan YN, Chiu HY, Lee FY, Yang JS and Tsai FJ: YC-1 induces G0/G1 phase arrest and mitochondria-dependent apoptosis in cisplatin-resistant human oral cancer CAR cells. Biomedicine (Taipei). 7(12)2017.PubMed/NCBI View Article : Google Scholar

32 

Yuan CH, Horng CT, Lee CF, Chiang NN, Tsai FJ, Lu CC, Chiang JH, Hsu YM, Yang JS and Chen FA: Epigallocatechin gallate sensitizes cisplatin-resistant oral cancer CAR cell apoptosis and autophagy through stimulating AKT/STAT3 pathway and suppressing multidrug resistance 1 signaling. Environ Toxicol. 32:845–855. 2017.PubMed/NCBI View Article : Google Scholar

33 

Chang HP, Lu CC, Chiang JH, Tsai FJ, Juan YN, Tsao JW, Chiu HY and Yang JS: Pterostilbene modulates the suppression of multidrug resistance protein 1 and triggers autophagic and apoptotic mechanisms in cisplatin-resistant human oral cancer CAR cells via AKT signaling. Int J Oncol. 52:1504–1514. 2018.PubMed/NCBI View Article : Google Scholar

34 

Lee HP, Chen PC, Wang SW, Fong YC, Tsai CH, Tsai FJ, Chung JG, Huang CY, Yang JS, Hsu YM, et al: Plumbagin suppresses endothelial progenitor cell-related angiogenesis in vitro and in vivo. J Funct Foods. 52:537–544. 2019.

35 

Chiang JH, Yang JS, Lu CC, Hour MJ, Chang SJ, Lee TH and Chung JG: Newly synthesized quinazolinone HMJ-38 suppresses angiogenetic responses and triggers human umbilical vein endothelial cell apoptosis through p53-modulated Fas/death receptor signaling. Toxicol Appl Pharmacol. 269:150–162. 2013.PubMed/NCBI View Article : Google Scholar

36 

Ma YS, Weng SW, Lin MW, Lu CC, Chiang JH, Yang JS, Lai KC, Lin JP, Tang NY, Lin JG and Chung JG: Antitumor effects of emodin on LS1034 human colon cancer cells in vitro and in vivo: Roles of apoptotic cell death and LS1034 tumor xenografts model. Food Chem Toxicol. 50:1271–1278. 2012.PubMed/NCBI View Article : Google Scholar

37 

Tsai YF, Chen YF, Hsiao CY, Huang CW, Lu CC, Tsai SC and Yang JS: Caspase dependent apoptotic death by gadolinium chloride (GdCl3) via reactive oxygen species production and MAPK signaling in rat C6 glioma cells. Oncol Rep. 41:1324–1332. 2019.PubMed/NCBI View Article : Google Scholar

38 

Chiu YJ, Hour MJ, Jin YA, Lu CC, Tsai FJ, Chen TL, Ma H, Juan YN and Yang JS: Disruption of IGF-1R signaling by a novel quinazoline derivative, HMJ-30, inhibits invasiveness and reverses epithelial-mesenchymal transition in osteosarcoma U-2 OS cells. Int J Oncol. 52:1465–1478. 2018.PubMed/NCBI View Article : Google Scholar

39 

Lu CC, Chiang JH, Tsai FJ, Hsu YM, Juan YN, Yang JS and Chiu HY: Metformin triggers the intrinsic apoptotic response in human AGS gastric adenocarcinoma cells by activating AMPK and suppressing mTOR/AKT signaling. Int J Oncol. 54:1271–1281. 2019.PubMed/NCBI View Article : Google Scholar

40 

Han J, Lee JD, Bibbs L and Ulevitch RJ: A MAP kinase targeted by endotoxin and hyperosmolarity in mammalian cells. Science. 265:808–811. 1994.PubMed/NCBI View Article : Google Scholar

41 

Zhang P, Li J, Tang X, Zhang J, Liang J and Zeng G: Dracorhodin perchlorate induces apoptosis in primary fibroblasts from human skin hypertrophic scars via participation of caspase-3. Eur J Pharmacol. 728:82–92. 2014.PubMed/NCBI View Article : Google Scholar

42 

Bowley E, O'Gorman DB and Gan BS: Beta-catenin signaling in fibroproliferative disease. J Surg Res. 138:141–150. 2007.PubMed/NCBI View Article : Google Scholar

43 

Lindley LE, Stojadinovic O, Pastar I and Tomic-Canic M: Biology and biomarkers for wound healing. Plast Reconstr Surg. 138 (Suppl 3):S18–S28. 2016.PubMed/NCBI View Article : Google Scholar

44 

Pereira Beserra F, Xue M, Maia GLA, Leite Rozza A, Helena Pellizzon C and Jackson CJ: Lupeol, a Pentacyclic Triterpene, promotes migration, wound closure, and contractile effect in vitro: Possible involvement of PI3K/Akt and p38/ERK/MAPK Pathways. Molecules. 23(2819)2018.PubMed/NCBI View Article : Google Scholar

45 

Patruno A, Pesce M, Grilli A, Speranza L, Franceschelli S, De Lutiis MA, Vianale G, Costantini E, Amerio P, Muraro R, et al: mTOR activation by PI3K/Akt and ERK signaling in short ELF-EMF exposed human keratinocytes. PLoS One. 10(e0139644)2015.PubMed/NCBI View Article : Google Scholar

46 

Bui NT, Ho MT, Kim YM, Lim Y and Cho M: Flavonoids promoting HaCaT migration: II. Molecular mechanism of 4',6,7-trimethoxyisoflavone via NOX2 activation. Phytomedicine. 21:570–577. 2014.PubMed/NCBI View Article : Google Scholar

47 

Yang HL, Tsai YC, Korivi M, Chang CT and Hseu YC: Lucidone promotes the Cutaneous Wound Healing process via activation of the PI3K/AKT, Wnt/β-catenin and NF-κB signaling pathways. Biochim Biophys Acta Mol Cell Res. 1864:151–168. 2017.PubMed/NCBI View Article : Google Scholar

48 

Lee SH, Zahoor M, Hwang JK, Min do S and Choi KY: Valproic acid induces cutaneous wound healing in vivo and enhances keratinocyte motility. PLoS One. 7(e48791)2012.PubMed/NCBI View Article : Google Scholar

49 

Yeh CJ, Chen CC, Leu YL, Lin MW, Chiu MM and Wang SH: The effects of artocarpin on wound healing: In vitro and in vivo studies. Sci Rep. 7(15599)2017.PubMed/NCBI View Article : Google Scholar

50 

Chen J, Chen Y, Chen Y, Yang Z, You B, Ruan YC and Peng Y: Epidermal CFTR Suppresses MAPK/NF-κB to Promote Cutaneous Wound Healing. Cell Physiol Biochem. 39:2262–2274. 2016.PubMed/NCBI View Article : Google Scholar

51 

Gazel A, Nijhawan RI, Walsh R and Blumenberg M: Transcriptional profiling defines the roles of ERK and p38 kinases in epidermal keratinocytes. J Cell Physiol. 215:292–308. 2008.PubMed/NCBI View Article : Google Scholar

52 

Saika S, Okada Y, Miyamoto T, Yamanaka O, Ohnishi Y, Ooshima A, Liu CY, Weng D and Kao WW: Role of p38 MAP kinase in regulation of cell migration and proliferation in healing corneal epithelium. Invest Ophthalmol Vis Sci. 45:100–109. 2004.PubMed/NCBI View Article : Google Scholar

53 

Sharma GD, He J and Bazan HE: p38 and ERK1/2 coordinate cellular migration and proliferation in epithelial wound healing: Evidence of cross-talk activation between MAP kinase cascades. J Biol Chem. 278:21989–21997. 2003.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2021
Volume 22 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu C, Yang J, Chiu Y, Tsai F, Hsu Y, Yin M, Juan Y, Ho T and Chen H: Dracorhodin perchlorate enhances wound healing via β‑catenin, ERK/p38, and AKT signaling in human HaCaT keratinocytes. Exp Ther Med 22: 822, 2021
APA
Lu, C., Yang, J., Chiu, Y., Tsai, F., Hsu, Y., Yin, M. ... Chen, H. (2021). Dracorhodin perchlorate enhances wound healing via β‑catenin, ERK/p38, and AKT signaling in human HaCaT keratinocytes. Experimental and Therapeutic Medicine, 22, 822. https://doi.org/10.3892/etm.2021.10254
MLA
Lu, C., Yang, J., Chiu, Y., Tsai, F., Hsu, Y., Yin, M., Juan, Y., Ho, T., Chen, H."Dracorhodin perchlorate enhances wound healing via β‑catenin, ERK/p38, and AKT signaling in human HaCaT keratinocytes". Experimental and Therapeutic Medicine 22.2 (2021): 822.
Chicago
Lu, C., Yang, J., Chiu, Y., Tsai, F., Hsu, Y., Yin, M., Juan, Y., Ho, T., Chen, H."Dracorhodin perchlorate enhances wound healing via β‑catenin, ERK/p38, and AKT signaling in human HaCaT keratinocytes". Experimental and Therapeutic Medicine 22, no. 2 (2021): 822. https://doi.org/10.3892/etm.2021.10254