Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2018, Vol. 12 Issue (3) : 289-300    https://doi.org/10.1007/s11684-017-0550-7
RESEARCH ARTICLE
Sub-cytotoxic concentrations of ionic silver promote the proliferation of human keratinocytes by inducing the production of reactive oxygen species
Xiaodong Duan1,2, Daizhi Peng1,3(), Yilan Zhang1, Yalan Huang1, Xiao Liu1, Ruifu Li1, Xin Zhou1, Jing Liu1
1. Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
2. Burn and Plastic Surgery Department, 209 Hospital of PLA, Mudanjiang 157011, China
3. Tissue Engineering Research Unit, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China
 Download: PDF(933 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Silver-containing preparations are widely used in the management of skin wounds, but the effects of silver ions on skin wound healing remain poorly understood. This study investigated the effects of silver ions (Ag+) on the proliferation of human skin keratinocytes (HaCaT) and the production of intracellular reactive oxygen species (ROS). After treating HaCaT cells with Ag+ and/or the active oxygen scavenger N-acetyl cysteine (NAC), cell proliferation and intracellular ROS generation were assessed using CCK-8 reagent and DCFH-DA fluorescent probe, respectively. In addition, 5-bromo-2-deoxyUridine (BrdU) incorporation assays, cell cycle flow cytometry, and proliferating cell nuclear antigen (PCNA) immunocytochemistry were conducted to further evaluate the effects of sub-cytotoxic Ag+ concentrations on HaCaT cells. The proliferation of HaCaT cells was promoted in the presence of 106 and 105 mol/L Ag+ at 24, 48, and 72 h. Intracellular ROS generation also significantly increased for 5–60 min after exposure to Ag+. The number of BrdU-positive cells and the presence of PCNA in HaCaT cells increased 48 h after the addition of 106 and 105 mol/L Ag+, with 105 mol/L Ag+ markedly increasing the cell proliferation index. These effects of sub-cytotoxic Ag+ concentrations were repressed by 5 mmol/L NAC. Our results suggest that sub-cytotoxic Ag+ concentrations promote the proliferation of human keratinocytes and might be associated with a moderate increase in intracellular ROS levels. This study provides important experimental evidence for developing novel silver-based wound agents or dressings with few or no cytotoxicity.

Keywords ionic silver      human keratinocyte      cell proliferation      reactive oxygen species      active oxygen scavenger      NAC     
Corresponding Author(s): Daizhi Peng   
Just Accepted Date: 04 August 2017   Online First Date: 03 November 2017    Issue Date: 04 May 2018
 Cite this article:   
Xiaodong Duan,Daizhi Peng,Yilan Zhang, et al. Sub-cytotoxic concentrations of ionic silver promote the proliferation of human keratinocytes by inducing the production of reactive oxygen species[J]. Front. Med., 2018, 12(3): 289-300.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-017-0550-7
https://academic.hep.com.cn/fmd/EN/Y2018/V12/I3/289
Fig.1  Ag+ influences the proliferation of HaCaT cells. (A–E) Relative proliferation ratios of HaCaT cells following treatment with various concentrations of Ag+. *P<0.05, **P<0.01, experimental versus negative control groups. (F) HaCaT cells (40× and 200×magnifications) following treatment with Ag+ for 48 h.
Fig.2  Effects of Ag+ on ROS generation in HaCaT cells. (A–F) Relative fluorescence intensity of DCFH-DA in HaCaT cells following treatment with Ag+. *P<0.05, **P<0.01 experimental versus negative control groups. (G) Representative images of HaCaT cells (400× magnification) after treatment with Ag+ for 48 h.
Fig.3  Effects of NAC on the proliferation of HaCaT cells treated with Ag+.
Fig.4  Effects of NAC and silver ion on ROS production in HaCaT cells. (A–F) Relative fluorescence intensity of intracellular DCFH-DA in HaCaT cells after exposure to Ag+ and NAC (n = 3). (G) Representative images of intracellular DCFH-DA 60 min after HaCaT cells were treated with Ag+ and NAC (400× magnification).
Fig.5  Effects of NAC and Ag+ on BrdU incorporation and PCNA expression in HaCaT cells. (A) Percentage of BrdU-positive cells reflecting the level of cell proliferation. (B) Immunofluorescent staining of PCNA. (C) Representative images (400× magnification) showing BrdU incorporation (red) and the presence of PCNA (green). Cell nuclei were stained with DAPI (blue).
Ag+ (mol/L)00106106105105
NAC (mmol/L)050505
G1 (%)48.23±5.5047.39±4.9647.49±5.9650.00±8.7039.91±2.23a50.15±6.72b
S (%)47.33±4.5048.42±4.8248.11±5.4145.82±5.3353.46±5.9646.84±6.32
G2 (%)4.44±4.564.20±1.074.41±3.184.19±4.866.64±7.963.01±3.67
PI51.77±5.4952.62±4.9652.52±5.9750.00±8.7060.10±2.23a 49.85±6.71b
Tab.1  Effects of NAC and Ag+ on cell cycle in HaCaT cells
Fig.6  Cell cycle distribution 48 h after treatment with Ag+ and NAC treatment. (A) 0 mol/L Ag+. (B) 105 mol/L Ag+. (C) 105 mol/L Ag+ and 5 mmol/L NAC.
1 Moyer CA, Brentano L, Gravens DL, Margraf HW Jr, Monafo WW Jr. Treatment of large human burns with 0.5 per cent silver nitrate solution. Arch Surg 1965; 90(6): 812–867
https://doi.org/10.1001/archsurg.1965.01320120014002 pmid: 14333527
2 Fox CL Jr. Silver sulfadiazine—a new topical therapy for Pseudomonas in burns. Therapy of Pseudomonas infection in burns. Arch Surg 1968; 96(2): 184–188
https://doi.org/10.1001/archsurg.1968.01330200022004 pmid: 5638080
3 Chopra I. The increasing use of silver-based products as antimicrobial agents: a useful development or a cause for concern? J Antimicrob Chemother 2007; 59(4): 587–590
https://doi.org/10.1093/jac/dkm006 pmid: 17307768
4 Swathy JR, Sankar MU, Chaudhary A, Aigal S, Anshup T, Pradeep. Antimicrobial silver: an unprecedented anion effect. Sci Rep 2014; 4: 7161
https://doi.org/10.1038/srep07161 pmid: 25418185
5 Mijnendonckx K, Leys N, Mahillon J, Silver S, Van Houdt R. Antimicrobial silver: uses, toxicity and potential for resistance. Biometals 2013; 26(4): 609–621
https://doi.org/10.1007/s10534-013-9645-z pmid: 23771576
6 Lansdown A, Williams A. Bacterial resistance to silver-based antibiotics. Nurs Times 2007; 103(9): 48–49
pmid: 17375724
7 Burd A, Kwok CH, Hung SC, Chan HS, Gu H, Lam WK, Huang L. A comparative study of the cytotoxicity of silver-based dressings in monolayer cell, tissue explant, and animal models. Wound Repair Regen 2007; 15(1): 94–104
https://doi.org/10.1111/j.1524-475X.2006.00190.x pmid: 17244325
8 Berger TJ, Spadaro JA, Chapin SE, Becker RO. Electrically generated silver ions: quantitative effects on bacterial and mammalian cells. Antimicrob Agents Chemother 1976; 9(2): 357–358
https://doi.org/10.1128/AAC.9.2.357 pmid: 944551
9 Arora S, Jain J, Rajwade JM, Paknikar KM. Interactions of silver nanoparticles with primary mouse fibroblasts and liver cells. Toxicol Appl Pharmacol 2009; 236(3): 310–318
https://doi.org/10.1016/j.taap.2009.02.020 pmid: 19269301
10 Zanette C, Pelin M, Crosera M, Adami G, Bovenzi M, Larese FF, Florio C. Silver nanoparticles exert a long-lasting antiproliferative effect on human keratinocyte HaCaT cell line. Toxicol In Vitro 2011; 25(5): 1053–1060
https://doi.org/10.1016/j.tiv.2011.04.005 pmid: 21501681
11 Poon VKM, Burd A. In vitro cytotoxity of silver: implication for clinical wound care. Burns 2004; 30(2): 140–147
https://doi.org/10.1016/j.burns.2003.09.030 pmid: 15019121
12 Morones-Ramirez JR, Winkler JA, Spina CS, Collins JJ. Silver enhances antibiotic activity against gram-negative bacteria. Sci Transl Med 2013; 5(190): 190ra81
https://doi.org/10.1126/scitranslmed.3006276 pmid: 23785037
13 Thomas GW, Rael LT, Bar-Or R, Shimonkevitz R, Mains CW, Slone DS, Craun ML, Bar-Or D. Mechanisms of delayed wound healing by commonly used antiseptics. J Trauma 2009, 66:82–90,  discussion 90–91
https://doi.org/10.1097/TA.0b013e31818b146d
14 Michaels JA, Campbell B, King B, Palfreyman SJ, Shackley P, Stevenson M. Randomized controlled trial and cost-effectiveness analysis of silver-donating antimicrobial dressings for venous leg ulcers (VULCAN trial). Br J Surg 2009; 96(10): 1147–1156
https://doi.org/10.1002/bjs.6786 pmid: 19787753
15 White R, Cutting K, Ousey K, Butcher M, Gray D, Flanagan M, Donnelly J, McIntosh C, Kingsley A, Fletcher J, Chadwick P, Gethin G, Beldon P. Randomized controlled trial and cost-effectiveness analysis of silver-donating antimicrobial dressings for venous leg ulcers (VULCAN trial) (Br J Surg 2009; 96: 1147-1156). Br J Surg 2010; 97(3): 459–460
https://doi.org/10.1002/bjs.7017 pmid: 20140943
16 Vermeulen H, van Hattem JM, Storm-Versloot MN, Ubbink DT. Topical silver for treating infected wounds. Cochrane Database Syst Rev 2007; (1): CD005486
pmid: 17253557
17 Storm-Versloot MN, Vos CG, Ubbink DT, Vermeulen H. Topical silver for preventing wound infection. Cochrane Database Syst Rev 2010; (3): CD006478
pmid: 20238345
18 Aziz Z, Abu SF, Chong NJ. A systematic review of silver-containing dressings and topical silver agents (used with dressings) for burn wounds. Burns 2012; 38(3): 307–318
https://doi.org/10.1016/j.burns.2011.09.020 pmid: 22030441
19 Gordon O, Vig Slenters T, Brunetto PS, Villaruz AE, Sturdevant DE, Otto M, Landmann R, Fromm KM. Silver coordination polymers for prevention of implant infection: thiol interaction, impact on respiratory chain enzymes, and hydroxyl radical induction. Antimicrob Agents Chemother 2010; 54(10): 4208– 4218
https://doi.org/10.1128/AAC.01830-09 pmid: 20660682
20 Foldbjerg R, Olesen P, Hougaard M, Dang DA, Hoffmann HJ, Autrup H. PVP-coated silver nanoparticles and silver ions induce reactive oxygen species, apoptosis and necrosis in THP-1 monocytes. Toxicol Lett 2009; 190(2): 156–162
https://doi.org/10.1016/j.toxlet.2009.07.009 pmid: 19607894
21 Miyayama T, Arai Y, Suzuki N, Hirano S. Mitochondrial electron transport is inhibited by disappearance of metallothionein in human bronchial epithelial cells following exposure to silver nitrate. Toxicology 2013; 305: 20–29
https://doi.org/10.1016/j.tox.2013.01.004 pmid: 23333424
22 Allen RG, Tresini M. Oxidative stress and gene regulation. Free Radic Biol Med 2000; 28(3): 463–499
https://doi.org/10.1016/S0891-5849(99)00242-7 pmid: 10699758
23 Yamamoto A, Honma R, Tanaka A, Sumita M. Generic tendency of metal salt cytotoxicity for six cell lines. J Biomed Mater Res 1999; 47(3): 396–403
https://doi.org/10.1002/(SICI)1097-4636(19991205)47:3<396::AID-JBM15>3.0.CO;2-R pmid: 10487892
24 Martin KR, Barrett JC. Reactive oxygen species as double-edged swords in cellular processes: low-dose cell signaling versus high-dose toxicity. Hum Exp Toxicol 2002; 21(2): 71–75
https://doi.org/10.1191/0960327102ht213oa pmid: 12102499
25 Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell 2012; 48(2): 158–167
https://doi.org/10.1016/j.molcel.2012.09.025 pmid: 23102266
26 Sauer H, Wartenberg M, Hescheler J. Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell Physiol Biochem 2001; 11(4): 173–186
https://doi.org/10.1159/000047804 pmid: 11509825
27 Samuni Y, Goldstein S, Dean OM, Berk M. The chemistry and biological activities of N-acetylcysteine. Biochim Biophys Acta. 2013;1830(8): 4117–4129
https://doi.org/10.1016/j.bbagen.2013.04.016
28 Bateman DN, Dear JW, Thanacoody HK, Thomas SH, Eddleston M, Sandilands EA, Coyle J, Cooper JG, Rodriguez A, Butcher I, Lewis SC, Vliegenthart AD, Veiraiah A, Webb DJ, Gray A. Reduction of adverse effects from intravenous acetylcysteine treatment for paracetamol poisoning: a randomised controlled trial. Lancet 2014; 383(9918): 697–704
https://doi.org/10.1016/S0140-6736(13)62062-0 pmid: 24290406
29 Tominaga H, Ishiyama M, Ohseto F, Sasamoto K, Hamamoto T, Suzuki K, Watanabe M. A water-soluble tetrazolium salt useful for colorimetric cell viability assay. Anal Commun 1999; 36(2): 47–50
https://doi.org/10.1039/a809656b
30 Bosq J, Bourhis J. Bromodeoxyuridine (BrdU). Analysis of cellular proliferation. Ann Pathol 1997; 17(3): 171–178 (in French)
pmid: 9296576
31 Chen AC, Arany PR, Huang YY, Tomkinson EM, Sharma SK, Kharkwal GB, Saleem T, Mooney D, Yull FE, Blackwell TS, Hamblin MR. Low-level laser therapy activates NF-κB via generation of reactive oxygen species in mouse embryonic fibroblasts. PLoS One 2011; 6(7): e22453
https://doi.org/10.1371/journal.pone.0022453 pmid: 21814580
32 Burdon RH. Superoxide and hydrogen peroxide in relation to mammalian cell proliferation. Free Radic Biol Med 1995; 18(4): 775–794
https://doi.org/10.1016/0891-5849(94)00198-S pmid: 7750801
33 Ruiz-Ginés JA, López-Ongil S, González-Rubio M, González-Santiago L, Rodrµguez-Puyol M, Rodrµguez-Puyol D. Reactive oxygen species induce proliferation of bovine aortic endothelial cells. J Cardiovasc Pharmacol 2000; 35(1): 109–113
https://doi.org/10.1097/00005344-200001000-00014 pmid: 10630740
34 Egelkrout EM, Mariconti L, Settlage SB, Cella R, Robertson D, Hanley-Bowdoin L. Two E2F elements regulate the proliferating cell nuclear antigen promoter differently during leaf development. Plant Cell 2002; 14(12): 3225–3236
https://doi.org/10.1105/tpc.006403 pmid: 12468739
35 Koundrioukoff S, Jónsson ZO, Hasan S, de Jong RN, van der Vliet PC, Hottiger MO, Hübscher U. A direct interaction between proliferating cell nuclear antigen (PCNA) and Cdk2 targets PCNA-interacting proteins for phosphorylation. J Biol Chem 2000; 275(30): 22882–22887
https://doi.org/10.1074/jbc.M001850200 pmid: 10930425
[1] Jing Ma, Shiyu Chen, Lili Hao, Wei Sheng, Weicheng Chen, Xiaojing Ma, Bowen Zhang, Duan Ma, Guoying Huang. Long non-coding RNA SAP30-2:1 is downregulated in congenital heart disease and regulates cell proliferation by targeting HAND2[J]. Front. Med., 2021, 15(1): 91-100.
[2] Jun Song, Yeping Huang, Wenjian Zheng, Jing Yan, Min Cheng, Ruxing Zhao, Li Chen, Cheng Hu, Weiping Jia. Resveratrol reduces intracellular reactive oxygen species levels by inducing autophagy through the AMPK-mTOR pathway[J]. Front. Med., 2018, 12(6): 697-706.
[3] Xinjian Li,Minna Cheng,Hao Zhang,Ting Ke,Yisheng Chen. Prevalence and determinations of physical inactivity among public hospital employees in Shanghai, China: a cross-sectional study[J]. Front. Med., 2015, 9(1): 100-107.
[4] Kai Qu,Ting Lin,Zhixin Wang,Sinan Liu,Hulin Chang,Xinsen Xu,Fandi Meng,Lei Zhou,Jichao Wei,Minghui Tai,Yafeng Dong,Chang Liu. Reactive oxygen species generation is essential for cisplatin-induced accelerated senescence in hepatocellular carcinoma[J]. Front. Med., 2014, 8(2): 227-235.
[5] Xiaowei GONG MD, PhD, Xiaoyan MING MD, Xu WANG MM, Daan WANG MD, Peng DENG MM, Yong JIANG MD, PhD, Aihua LIU MD, PhD, . Effect of PRAK gene knockout on the proliferation of mouse embryonic fibroblasts[J]. Front. Med., 2009, 3(4): 379-383.
[6] Qigong LIU, Honglian ZHOU, Yan ZENG, Shan YE, Jiani LIU, Zaiying LU. Mechanism of vascular endothelial growth factor on the prevention of restenosis after angioplasty[J]. Front Med Chin, 2009, 3(2): 177-180.
[7] Gang WANG. NADPH oxidase and reactive oxygen species as signaling molecules in carcinogenesis[J]. Front Med Chin, 2009, 3(1): 1-7.
[8] JIN Qiumei, LI Yan, SUN Zengrong. Estrogenic activities of di-2-ethylhexyl phthalate[J]. Front. Med., 2008, 2(3): 303-308.
[9] ZOU Yunfeng, NIU Piye, GONG Zhiyong, YANG Jin, YUAN Jing, WU Tangchun, CHEN Xuemin. Relationship between reactive oxygen species and sodium-selenite-induced DNA damage in HepG2 cells[J]. Front. Med., 2007, 1(3): 327-332.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed