DOI QR코드

DOI QR Code

Beneficial effects of Diplectria barbata (Wall. Ex C. B. Clarke) Franken et Roos extract on aging and antioxidants in vitro and in vivo

  • Hong, Youngeun (Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University) ;
  • Lee, Hyunji (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) ;
  • Bayarmunkh, Choinyam (Department of Graduate Education, Graduate school, Mongolian National University of Medical Sciences) ;
  • Boldbaatar, Damdindorj (Department of Graduate Education, Graduate school, Mongolian National University of Medical Sciences) ;
  • Kwon, So Hee (College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei 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 : 2020.07.03
  • Accepted : 2020.09.01
  • Published : 2021.01.15

Abstract

The purpose of this study is to explore the effects of Diplectria barbata (Wall. Ex C.B. Clarke) Franken & Roons (DFR) on wound healing, antioxidant and aging in Normal Human Dermal Fibroblast cell (NHDF) cells and mouse skin models. We investigated the effects of the aging process in vitro and in vivo. DFRtreated NHDF cells showed a concentration-dependent increase in the expression of extracellular matrix (ECM) proteins (Collagen-2.5-fold increase at 50 ㎍/ml, Elastin-1.5-fold increase at 1㎍/ml) as well as an increase in proteins related to cell survival, differentiation, and development, while expression of aging proteins such as matrix metalloproteinase 3 (MMP-3) was decreased (5-fold decrease at 50 ㎍/ml). DFR treatment also led to enhanced expression of antioxidant proteins such as nuclear factor erythroid 2-related factor 2 (10-fold increase at 50 ㎍/ml) and heme oxygenase 1 (1.5-fold increase at 25 ㎍/ml). To further investigate the antioxidative effects of DFR extracts, the 2,2-diphenyl-1-picrylhydrazyl radical scavenging activities were also evaluated. DFR extracts improved wound healing and resulted in increased expression of ECM proteins, while enzymes involved in collagen degradation, including MMP-3, were decreased in NHDF cells as well as in a mouse model. This study demonstrates the anti-aging, antioxidant, and wound healing properties of DFR extracts. Therefore, DFR extracts present may facilitate skin protection and care.

Keywords

Acknowledgement

We would like to thank KRIBB (Korea Research Institute of bioscience & Biotechnology) for providing to DFR Extracts. This work was financially supported by a research fund from Chungnam National University (grant to J. Park) and by the Brain Korea 21 PLUS Project for Medical Science, Chungnam National University School of Medicine. The English in this document has been checked by at least two professional editors, both native speakers of English. For a certificate, please see: http://www.textcheck.com/certificate/LL0V6r.

References

  1. You J et al (2015) The antiaging properties of Andrographis paniculata by activation epidermal cell stemness. Molecules 20:17557-17569. https://doi.org/10.3390/molecules200917557
  2. Masaki H (2010) Role of antioxidants in the skin: anti-aging effects. J Dermatol Sci 58:85-90. https://doi.org/10.1016/j.jdermsci.2010.03.003
  3. Teti A (1992) Regulation of cellular functions by extracellular matrix. J Am Soc Nephrol 2:S83-S87 https://doi.org/10.1681/ASN.V210s83
  4. Di Lullo GA et al (2002) Mapping the ligand-binding sites and disease-associated mutations on the most abundant protein in the human, type I collagen. J Biol Chem 277:4223-4231. https://doi.org/10.1074/jbc.M110709200
  5. Song G, Ouyang G, Bao S (2005) The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med 9:59-71. https://doi.org/10.1111/j.1582-4934.2005.tb00337.x
  6. Pearson G et al (2001) Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 22:153-183. https://doi.org/10.1210/edrv.22.2.0428
  7. Woodgett JR, Avruch J, Kyriakis J (1996) The stress activated protein kinase pathway. Cancer Surv 27:127-138. https://doi.org/10.1210/edrv.22.2.0428
  8. Stephens L et al (1998) Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B. Science 279:710-714. https://doi.org/10.1126/science.279.5351.710
  9. Purdom-Dickinson SE et al (2007) 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 42:159-176. https://doi.org/10.1016/j.yjmcc.2006.09.012
  10. Zhu H et al (2005) 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 579:3029-3036. https://doi.org/10.1016/j.febslet.2005.04.058
  11. Li W et al (2008) Activation of Nrf2-antioxidant signaling attenuates NFkappaB-inflammatory response and elicits apoptosis. Biochem Pharmacol 76:1485-1489. https://doi.org/10.1016/j.bcp.2008.07.017
  12. Rinnerthaler M et al (2015) Oxidative stress in aging human skin. Biomolecules 5:545-589. https://doi.org/10.3390/biom5020545
  13. Su ZY et al (2013) A perspective on dietary phytochemicals and cancer chemoprevention: oxidative stress, nrf2, and epigenomics. Top Curr Chem 329:133-162. https://doi.org/10.1007/128_2012_340
  14. Surh YJ (2003) Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer 3:768-780. https://doi.org/10.1038/nrc1189
  15. Lee H et al (2016) 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 11:1017-1026. https://doi.org/10.2147/CIA.S107734
  16. Park J et al (2017) Recognition of transmembrane protein 39A as a tumor-specific marker in brain tumor. Toxicol Res 33:63-69. https://doi.org/10.5487/TR.2017.33.1.063
  17. Westermarck J, Kahari VM (1999) Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 13:781-792. https://doi.org/10.1096/fasebj.13.8.781
  18. Romagna-Genon C (2001) Comparative clinical study of guided tissue regeneration with a bioabsorbable bilayer collagen membrane and subepithelial connective tissue graft. J Periodontol 72:1258-1264. https://doi.org/10.1902/jop.2000.72.9.1258
  19. Popescu MC et al (2010) Biomaterials based on new polyurethane and hydrolyzed collagen, k-elastin, hyaluronic acid and chondroitin sulfate. Int J Biol Macromol 47:646-653. https://doi.org/10.1016/j.ijbiomac.2010.08.013
  20. Xue M, Jackson CJ (2015) Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care (New Rochelle) 4:119-136. https://doi.org/10.1089/wound.2013.0485
  21. Banik D et al (2015) MMP3-mediated tumor progression is controlled transcriptionally by a novel IRF8-MMP3 interaction. Oncotarget 6:15164-15179. https://doi.org/10.18632/oncotarget.3897
  22. Rahal A et al (2014) Oxidative stress, prooxidants, and antioxidants: the interplay. Biomed Res Int 2014:761264. https://doi.org/10.1155/2014/761264
  23. Hanson KM, Clegg RM (2002) Observation and quantification of ultraviolet-induced reactive oxygen species in ex vivo human skin. Photochem Photobiol 76:57-63. https://doi.org/10.1562/0031-8655(2002)076<0057:oaqoui>2.0.co;2
  24. Sakurai H et al (2005) 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 4:715-720. https://doi.org/10.1039/b417319h
  25. Bickers DR, Athar M (2006) Oxidative stress in the pathogenesis of skin disease. J Invest Dermatol 126:2565-2575. https://doi.org/10.1038/sj.jid.5700340
  26. Briganti S, Picardo M (2003) Antioxidant activity, lipid peroxidation and skin diseases. What's new. J Eur Acad Dermatol Venereol 17:663-669. https://doi.org/10.1046/j.1468-3083.2003.00751.x
  27. Almeida IF et al (2015) Protective effect of C. sativa leaf extract against UV mediated-DNA damage in a human keratinocyte cell line. J Photochem Photobiol B 144:28-34. https://doi.org/10.1016/j.jphotobiol.2015.01.010
  28. Hseu YC et al (2015) Dermato-protective properties of ergothioneine through induction of Nrf2/ARE-mediated antioxidant genes in UVA-irradiated human keratinocytes. Free Radic Biol Med 86:102-117. https://doi.org/10.1016/j.freeradbiomed.2015.05.026
  29. Radjendirane V et al (1998) Disruption of the DT diaphorase (NQO1) gene in mice leads to increased menadione toxicity. J Biol Chem 273:7382-7389. https://doi.org/10.1074/jbc.273.13.7382
  30. Srisook K, Kim C, Cha YN (2005) Molecular mechanisms involved in enhancing HO-1 expression: de-repression by heme and activation by Nrf2, the "one-two" punch. Antioxid Redox Signal 7:1674-1687. https://doi.org/10.1089/ars.2005.7.1674
  31. Woodford FP, Whitehead TP (1998) Is measuring serum antioxidant capacity clinically useful? Ann Clin Biochem 35:48-56. https://doi.org/10.1177/000456329803500105
  32. Seo MY et al (2009) Anti-aging effect of rice wine in cultured human fibroblasts and keratinocytes. J Biosci Bioeng 107:266-271. https://doi.org/10.1016/j.jbiosc.2008.11.016
  33. Krafts KP (2010) Tissue repair: the hidden drama. Organogenesis 6:225-233. https://doi.org/10.4161/org.6.4.12555
  34. Guo S, Dipietro LA (2010) Factors affecting wound healing. J Dent Res 89:219-229. https://doi.org/10.1177/0022034509359125
  35. Apak R et al (2007) Comparative evaluation of various total antioxidant capacity assays applied to phenolic compounds with the CUPRAC assay. Molecules 12:1496-1547. https://doi.org/10.3390/12071496
  36. Jere SW, Houreld NN, Abrahamse H (2019) Role of the PI3K/AKT (mTOR and GSK3beta) signalling pathway and photobiomodulation in diabetic wound healing. Cytokine Growth Factor Rev 50:52-59. https://doi.org/10.1016/j.cytogfr.2019.03.001
  37. Huang H et al (2015) Impaired wound healing results from the dysfunction of the Akt/mTOR pathway in diabetic rats. J Dermatol Sci 79:241-251. https://doi.org/10.1016/j.jdermsci.2015.06.002
  38. Coussens LM, Werb Z (2002) Inflammation cancer. Nature 420:860-867. https://doi.org/10.1038/nature01322
  39. Chen L et al (2008) Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 3:e1886. https://doi.org/10.1371/journal.pone.0001886

Cited by

  1. Betulinic Acid Restricts Human Bladder Cancer Cell Proliferation In Vitro by Inducing Caspase-Dependent Cell Death and Cell Cycle Arrest, and Decreasing Metastatic Potential vol.26, pp.5, 2021, https://doi.org/10.3390/molecules26051381