Liquid plasma as a treatment for cutaneous wound healing through regulation of redox metabolism

Hye Ran Lee, Sung Un Kang, Haeng Jun Kim, Eun Jong Ji, Ju Hyun Yun, Sungryeal Kim, Jeon Yeob Jang, Yoo Seob Shin, Chul-Ho Kim, Hye Ran Lee, Sung Un Kang, Haeng Jun Kim, Eun Jong Ji, Ju Hyun Yun, Sungryeal Kim, Jeon Yeob Jang, Yoo Seob Shin, Chul-Ho Kim

Abstract

The skin functions as the outermost protective barrier to the internal organs and major vessels; thus, delayed regeneration from acute injury could induce serious clinical complications. For rapid recovery of skin wounds, promoting re-epithelialization of the epidermis at the initial stage of injury is essential, wherein epithelial keratinocytes act as leading cells via migration. This study applied plasma technology, which has been known to enable wound healing in the medical field. Through in vitro and in vivo experiments, the study elucidated the effect and molecular mechanism of the liquid plasma (LP) manufactured by our microwave plasma system, which was found to improve the applicability of existing gas-type plasma on skin cell migration for re-epithelialization. LP treatment promoted the cytoskeletal transformation of keratinocytes and migration owing to changes in the expression of integrin-dependent focal adhesion molecules and matrix metalloproteinases (MMPs). This study also identified the role of increased levels of intracellular reactive oxygen species (ROS) as a driving force for cell migration activation, which was regulated by changes in NADPH oxidases and mitochondrial membrane potential. In an in vivo experiment using a murine dorsal full-thickness acute skin wound model, LP treatment helped improve the re-epithelialization rate, reaffirming the activation of the underlying intracellular ROS-dependent integrin-dependent signaling molecules. These findings indicate that LP could be a valuable wound management material that can improve the regeneration potential of the skin via the activation of migration-related molecular signaling within the epithelial cell itself with plasma-driven oxidative eustress.

Conflict of interest statement

The authors declare no competing interests.

© 2023. The Author(s).

Figures

Fig. 1. Effects of LP treatment on…
Fig. 1. Effects of LP treatment on cell migration.
A Scratch wound migration assay after LP treatment on HaCaT (human keratinocyte) cells. The cells were plated in six-well plates, grown to 90% confluency, and with a monolayer denuded with a sterile pipette tip. Scratch wound migrations (scale bars = 500 μm) were documented by photography after 24 h of incubation. The data graph presents the mean ± standard deviation of three independent experiments. *P < 0.05, **P < 0.01, and ***P < .001. B SEM analysis (scale bars = 10 μm) of morphological changes in cytoskeletal (red indicated) structure in HaCaT cells that were either left untreated or treated with various diluted LP for 24 h. C Gelatin zymography for matrix metalloproteinase (MMP) in HaCaT cells. LP increased MMP-2 enzyme activity. Bar graph presents the mean ± standard deviation of three independent experiments. ***P < 0.001. D Quantitative PCR analysis was used for the expression level of MMP-2 in HaCaT cells. The mRNA expression of MMP-2 was significantly increased in the group treated with LP. Asterisks indicate statistically significant differences (***P < 0.001).
Fig. 2. LP increases the expression of…
Fig. 2. LP increases the expression of proteins involved in extracellular matrix (ECM) signaling.
A Expression of proteins involved in ECM signaling in HaCaT cells was confirmed by western blot. The degree of protein expression of integrin β1, p-FAK, and p-paxillin was significantly increased with LP (1/4) treatment time than the control group. All the western blotting experiments were performed under the same condition. After transferring the blots onto PVDF membranes, we immediately cropped the targeted blots according to referenced indicating markers. Targeted proteins were immunoblotted with the specific antibody for protein normalization. B Integrin β1 protein levels were determined by immunocytochemistry after LP (1/4) treatment for 24 h. Representative fluorescence microscopic images of HaCaT cells are shown. Scale bars = 50 μm. The increase in the expression of ECM molecule after LP treatment was confirmed using immunofluorescence assay. The green-stained stock in the cytoplasm represents p-FAK (C), and p-paxillin (D). The amount of green-stained stock increased in the LP-treated group. We performed Texas-Red conjugated phalloidin to visualize the cytoskeleton (F-actin) and used Hoechst 33258 to label cell nuclei. Each figure is representative of three experiments with triplicates.
Fig. 3. Treatment of LP increases cellular…
Fig. 3. Treatment of LP increases cellular oxidative stress and mitochondrial membrane potential (∆Ψm).
A The HaCaT cells treated with LP for 24 h were analyzed using flow cytometry, with dihydroethidium and 5-(and-6)-carboxy-2’,7’-dichlorodihydrofluorescein diacetate staining. B For the measurement of mitochondrial superoxide, the cells were incubated with 2.5 mM of MitoSOX and then stained and analyzed using flow cytometry. In (A) and (B), the data graph presents the mean ± standard deviation of three independent experiments. ***P < 0.001. C Measurement of ∆Ψm with JC-1. The JC-1 fluorescence shifted from red-orange to green, indicating depolarization of the ∆Ψm. The ∆Ψm change was measured objectively using a fluorescence microscope and flow cytometry. Bar graph presents the mean ± standard deviation of three independent experiments, calculated as a percentage of the control. **P < 0.01, ***P < 0.001. D Intracellular ROS stimulated with LP induces expression of NOX3 protein in HaCaT cells. Western blotting analysis for NOX3 antibody. GAPDH was used as the loading control.
Fig. 4. Correlation among LP-induced cellular ROS…
Fig. 4. Correlation among LP-induced cellular ROS generation and ECM signaling activation.
The HaCaT cells were incubated for 1 h before treatment with LP in the presence or absence of NAC (10 mM). A Scratch wound migration assay after LP treatment on HaCaT (human keratinocyte) cells. LP significantly increased cell migration across the cell stripped area, but significantly attenuated it when treated with the ROS scavenger, NAC. B Gelatin zymography for MMP-2. NAC attenuated the enzymatic activity of MMP-2 increased by LP. Bar graphs represent the mean ± standard deviation of three independent experiments. ***P < 0.001. C Quantitative PCR for MMP-2. The mRNA levels of MMP-2 decreased after NAC treatment. Each figure is representative of three experiments with triplicates. **P < 0.01, ***P < 0.001. D, B SEM images confirmed the effect of NTP on cell morphology. The cytoplasm of LP-treated HaCaT cells showed an increase in horizontal polarization and contraction and cytoplasmic protrusion compared with the control group (red arrow), but it was significantly inhibited in NAC-treated cells. E Western blot analysis. Integrin β1,3,5, p-FAK, and p-paxillin expression were suppressed significantly after NAC treatment to HaCaT cells. FH Immunofluorescence analysis confirmed whether NAC could attenuate the expression of integrin β1 (F) and ECM molecules (p-FAK (G), and p-paxillin (H)) increased by LP. We performed Texas-Red conjugated phalloidin to visualize the cytoskeleton (F-actin); we used Hoechst 33258 to label cell nuclei. Each figure is representative of three experiments with triplicates. Scale bars = 50 μm.
Fig. 5. LP treatment promoted wound regeneration…
Fig. 5. LP treatment promoted wound regeneration of murine dorsal acute skin wounds by activating intracellular ROS and epithelial cell adhesion molecules.
Twenty rats were separated into untreated control (n = 10) or LP treatment (n = 10) groups. A Images represent control and LP-treated wounds in rat dorsum. Two full-thickness wounds, 8 mm in diameter, were created on both sides of a rat’s upper back. Left: photographs of both sides of control and LP-treated (upper) and PBS (control) or LP-soaked gauze applied wounds (lower). Right: photograph of initially created wound on day 1 and its cross-section histologic image. Scale bar = 3000 μm. B Photographs of dorsal skin wound closures (left), which were controlled and treated with LP on days 1, 3, 7, 11, and 14. Quantification of the relative wound area (%) to the initial wound in control and LP-treated wounds (right). Mean ± SEM, *P < 0.05, **P < 0.01. C Left: On days 7 and 14 after wound creation, the wounds’ cross-sections stained by H&E staining showed decreased wound diameter in the LP-treated group. Ki-67 expressions were detected by IHC in each wound epithelium (black squares) and demonstrated increased expression in the basal layer of the epithelium in the LP-treated group. Scale bar = 3000 μm (H&E images)/100 μm (IHC images). Right: Positive immunostaining area of Ki-67 in both control and LP-treated groups on days 7 and 14 represented graphically. Mean ± SEM. D Left: To evaluate the effect of LP treatment on wound tissues, we performed NOX3, integrin β1, p-FAK, and p-paxillin IHC staining in both control and LP-treated groups on days 7 and 14. The IHC images in each box are 10× the magnification of the star-marked area in inserted H&E images of each box with an original magnification of 40×. Scale bar = 100 μm (IHC images), 3000 μm (H&E images). Right: Positive immunostaining area of each marker in both control and LP-treated groups on days 7 and 14 represented graphically. Mean ± SEM, *P < 0.05, **P < 0.01, and ***P < 0.001. E Schematic of LP treatment on keratinocytes and intracellular ROS mediated adhesion molecule activation as a subsequent process for promoting cell migration and wound regeneration.

References

    1. Dreifke MB, Jayasuriya AA, Jayasuriya AC. Current wound healing procedures and potential care. Mater Sci Eng C Mater Biol Appl. 2015;48:651–62. doi: 10.1016/j.msec.2014.12.068.
    1. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr266. doi: 10.1126/scitranslmed.3009337.
    1. Zhong SP, Zhang YZ, Lim CT. Tissue scaffolds for skin wound healing and dermal reconstruction. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2:510–25. doi: 10.1002/wnan.100.
    1. Vowden K, Vowden P. Wound dressings: principles and practice. Surgery. 2017;35:489–94.
    1. Bishop SM, Walker M, Rogers AA, Chen WY. Importance of moisture balance at the wound-dressing interface. J Wound Care. 2003;12:125–8. doi: 10.12968/jowc.2003.12.4.26484.
    1. Rodeheaver GT, Ratliff CR. Chronic wound care: a clinical source book for healthcare professionals. Wayne, PA: Health Management Publications; 1997. p. 97–108.
    1. Atiyeh BS, Dibo SA, Hayek SN. Wound cleansing, topical antiseptics and wound healing. Int Wound J. 2009;6:420–30. doi: 10.1111/j.1742-481X.2009.00639.x.
    1. Wilkins RG, Unverdorben M. Wound cleaning and wound healing: a concise review. Adv Ski Wound Care. 2013;26:160–3. doi: 10.1097/01.ASW.0000428861.26671.41.
    1. Tottoli EM, Dorati R, Genta I, Chiesa E, Pisani S, Conti B. Skin wound healing process and new emerging technologies for skin wound care and regeneration. Pharmaceutics. 2020;12:735.
    1. Wilkinson HN, Hardman MJ. Wound healing: cellular mechanisms and pathological outcomes. Open Biol. 2020;10:200223. doi: 10.1098/rsob.200223.
    1. Raziyeva K, Kim Y, Zharkinbekov Z, Kassymbek K, Jimi S, Saparov A. Immunology of acute and chronic wound healing. Biomolecules. 2021;11:700.
    1. Vonw T, Schmidt A, Bekeschus S, Wende K, Weltmann KD. Plasma medicine: a field of applied redox biology. In Vivo. 2019;33:1011–26. doi: 10.21873/invivo.11570.
    1. von Woedtke T, Reuter S, Masur K, Weltmann KD. Plasmas for medicine. Phys Rep. 2013;530:291–320. doi: 10.1016/j.physrep.2013.05.005.
    1. Sakudo A, Yagyu Y, Onodera T. Disinfection and sterilization using plasma technology: fundamentals and future perspectives for biological applications. Int J Mol Sci. 2019;20:5216.
    1. Boeckmann L, Schäfer M, Bernhardt T, Semmler ML, Jung O, Ojak G, et al. Cold atmospheric pressure plasma in wound healing and cancer treatment. Appl Sci. 2020;10:6898.
    1. Joseph A, Rane R, Vaid AJWHR. Atmospheric pressure plasma therapy for wound healing and disinfection: a review. In: Kumar, P., Kothari, V. (eds) Wound Healing Research. Springer, Singapore. 2021. p. 621–41.
    1. Kang SU, Choi JW, Chang JW, Kim KI, Kim YS, Park JK, et al. N(2) non-thermal atmospheric pressure plasma promotes wound healing in vitro and in vivo: potential modulation of adhesion molecules and matrix metalloproteinase-9. Exp Dermatol. 2017;26:163–70. doi: 10.1111/exd.13229.
    1. Choi JW, Kang SU, Kim YE, Park JK, Yang SS, Kim YS, et al. Novel therapeutic effects of non-thermal atmospheric pressure plasma for muscle regeneration and differentiation. Sci Rep. 2016;6:28829. doi: 10.1038/srep28829.
    1. Park JK, Kim YS, Kang SU, Lee YS, Won H-R, Kim C-H. Nonthermal atmospheric plasma enhances myoblast differentiation by eliciting STAT3 phosphorylation. FASEB J. 2019;33:4097–106.
    1. Schmidt A, Liebelt G, Nießner F, von Woedtke T, Bekeschus S. Gas plasma-spurred wound healing is accompanied by regulation of focal adhesion, matrix remodeling, and tissue oxygenation. Redox Biol. 2021;38:101809. doi: 10.1016/j.redox.2020.101809.
    1. Schmidt A, Bekeschus S, Wende K, Vollmar B, von Woedtke T. A cold plasma jet accelerates wound healing in a murine model of full-thickness skin wounds. Exp Dermatol. 2017;26:156–62. doi: 10.1111/exd.13156.
    1. Zhou R, Zhou R, Wang P, Xian Y, Mai-Prochnow A, Lu X, et al. Plasma-activated water: generation, origin of reactive species and biological applications. J Phys D: Appl Phys. 2020;53:303001.
    1. Bruggeman P, Kushner MJ, Locke BR, Gardeniers JG, Graham W, Graves DB, et al. Plasma–liquid interactions: a review and roadmap. Plasma Sources Sci Technol. 2016;25:053002.
    1. Thirumdas R, Kothakota A, Annapure U, Siliveru K, Blundell R, Gatt R, et al. Plasma activated water (PAW): chemistry, physico-chemical properties, applications in food and agriculture. Trends Food Sci Technol. 2018;77:21–31.
    1. Wang S, Xu D, Qi M, Li B, Peng S, Li Q, et al. Plasma-activated water promotes wound healing by regulating inflammatory responses. Biophysica. 2021;1:297–310.
    1. Xu D, Wang S, Li B, Qi M, Feng R, Li Q, et al. Effects of plasma-activated water on skin wound healing in mice. Microorganisms. 2020;8:1091.
    1. Hong YC, Ma SH, Kim K, Shin YW. Multihole dielectric barrier discharge with asymmetric electrode arrangement in water and application to sterilization of aqua pathogens. Chem Eng J. 2019;374:133–43. doi: 10.1016/j.cej.2019.05.178.
    1. Kotamraju S, Hogg N, Joseph J, Keefer LK, Kalyanaraman B. Inhibition of oxidized low-density lipoprotein-induced apoptosis in endothelial cells by nitric oxide. Peroxyl radical scavenging as an antiapoptotic mechanism. J Biol Chem. 2001;276:17316–23. doi: 10.1074/jbc.M011731200.
    1. Agren MS. Gelatinase activity during wound healing. Br J Dermatol. 1994;131:634–40. doi: 10.1111/j.1365-2133.1994.tb04974.x.
    1. Caley MP, Martins VLC, O’Toole EA. Metalloproteinases and wound healing. Adv Wound Care. 2015;4:225–34. doi: 10.1089/wound.2014.0581.
    1. Hopkinson SB, Hamill KJ, Wu Y, Eisenberg JL, Hiroyasu S, Jones JCR. Focal contact and hemidesmosomal proteins in keratinocyte migration and wound repair. Adv Wound Care. 2014;3:247–63. doi: 10.1089/wound.2013.0489.
    1. Lü D, Li Z, Gao Y, Luo C, Zhang F, Zheng L, et al. β1 integrin signaling in asymmetric migration of keratinocytes under mechanical stretch in a co-cultured wound repair model. Biomed Eng Online. 2016;15:130. doi: 10.1186/s12938-016-0263-1.
    1. Kang SU, Seo SJ, Kim YS, Shin YS, Koh YW, Lee CM, et al. Comparative effects of non-thermal atmospheric pressure plasma on migration and invasion in oral squamous cell cancer, by gas type. Yonsei Med J. 2017;58:272–81. doi: 10.3349/ymj.2017.58.2.272.
    1. Schaks M, Giannone G, Rottner K. Actin dynamics in cell migration. Essays Biochem. 2019;63:483–95. doi: 10.1042/EBC20190015.
    1. Graves DB. The emerging role of reactive oxygen and nitrogen species in redox biology and some implications for plasma applications to medicine and biology. J Phys D: Appl Phys. 2012;45:263001. doi: 10.1088/0022-3727/45/26/263001.
    1. Haertel B, von Woedtke T, Weltmann KD, Lindequist U. Non-thermal atmospheric-pressure plasma possible application in wound healing. Biomol Ther. 2014;22:477–90. doi: 10.4062/biomolther.2014.105.
    1. Chiarugi P, Pani G, Giannoni E, Taddei M, Colavitti R, Raugei G, et al. Reactive oxygen species as essential mediators of cell adhesion: the oxidative inhibition of a FAK tyrosine phosphatase is required for cell adhesion. J Cell Biol. 2003;161:933–44. doi: 10.1083/jcb.200211118.
    1. Svineng G, Ravuri C, Rikardsen O, Huseby NE, Winberg JO. The role of reactive oxygen species in integrin and matrix metalloproteinase expression and function. Connect Tissue Res. 2008;49:197–202. doi: 10.1080/03008200802143166.
    1. Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev. 2014;94:909–50. doi: 10.1152/physrev.00026.2013.
    1. Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21:363–83. doi: 10.1038/s41580-020-0230-3.
    1. Zorova LD, Popkov VA, Plotnikov EY, Silachev DN, Pevzner IB, Jankauskas SS, et al. Mitochondrial membrane potential. Anal Biochem. 2018;552:50–9. doi: 10.1016/j.ab.2017.07.009.
    1. Jiang F, Zhang Y, Dusting GJ. NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev. 2011;63:218–42. doi: 10.1124/pr.110.002980.
    1. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87:245–313. doi: 10.1152/physrev.00044.2005.
    1. Rezaie F, Momeni-Moghaddam M, Naderi-Meshkin H. Regeneration and repair of skin wounds: various strategies for treatment. Int J Low Extremity Wounds. 2019;18:247–61. doi: 10.1177/1534734619859214.
    1. Demidova-Rice TN, Hamblin MR, Herman IM. Acute and impaired wound healing: pathophysiology and current methods for drug delivery, part 1: normal and chronic wounds: biology, causes, and approaches to care. Adv Ski Wound Care. 2012;25:304–14. doi: 10.1097/01.ASW.0000416006.55218.d0.
    1. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314–21. doi: 10.1038/nature07039.
    1. Lü D, Liu X, Gao Y, Huo B, Kang Y, Chen J, et al. Asymmetric migration of human keratinocytes under mechanical stretch and cocultured fibroblasts in a wound repair model. PloS One. 2013;8:e74563. doi: 10.1371/journal.pone.0074563.
    1. Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173:370–8. doi: 10.1111/bjd.13954.
    1. Raja, Sivamani K, Garcia MS, Isseroff RR. Wound re-epithelialization: modulating keratinocyte migration in wound healing. Front Biosci: J Virtual Libr. 2007;12:2849–68. doi: 10.2741/2277.
    1. Pastar I, Stojadinovic O, Yin NC, Ramirez H, Nusbaum AG, Sawaya A, et al. Epithelialization in wound healing: a comprehensive review. Adv Wound Care. 2014;3:445–64. doi: 10.1089/wound.2013.0473.
    1. Coulombe PA. Towards a molecular definition of keratinocyte activation after acute injury to stratified epithelia. Biochem Biophys Res Commun. 1997;236:231–8. doi: 10.1006/bbrc.1997.6945.
    1. Longmate WM, Dipersio CM. Integrin regulation of epidermal functions in wounds. Adv Wound Care. 2014;3:229–46. doi: 10.1089/wound.2013.0516.
    1. Schmidt A, Liebelt G, Niessner F, von Woedtke T, Bekeschus S. Gas plasma-spurred wound healing is accompanied by regulation of focal adhesion, matrix remodeling, and tissue oxygenation. Redox Biol. 2021;38:101809. doi: 10.1016/j.redox.2020.101809.
    1. Schiller HB, Hermann MR, Polleux J, Vignaud T, Zanivan S, Friedel CC, et al. beta1- and alphav-class integrins cooperate to regulate myosin II during rigidity sensing of fibronectin-based microenvironments. Nat Cell Biol. 2013;15:625–36. doi: 10.1038/ncb2747.
    1. Grose R, Hutter C, Bloch W, Thorey I, Watt FM, Fässler R, et al. A crucial role of beta 1 integrins for keratinocyte migration in vitro and during cutaneous wound repair. Development. 2002;129:2303–15. doi: 10.1242/dev.129.9.2303.
    1. Brakebusch C, Fässler R. beta 1 integrin function in vivo: adhesion, migration and more. Cancer Metastasis Rev. 2005;24:403–11. doi: 10.1007/s10555-005-5132-5.
    1. Sieg DJ, Hauck CR, Schlaepfer DD. Required role of focal adhesion kinase (FAK) for integrin-stimulated cell migration. J Cell Sci. 1999;112:2677–91. doi: 10.1242/jcs.112.16.2677.
    1. Lopez-Colome AM, Lee-Rivera I, Benavides-Hidalgo R, Lopez E. Paxillin: a crossroad in pathological cell migration. J Hematol Oncol. 2017;10:50. doi: 10.1186/s13045-017-0418-y.
    1. Tan F, Rui X, Xiang X, Yu Z, Al-Rubeai M. Multimodal treatment combining cold atmospheric plasma and acidic fibroblast growth factor for multi-tissue regeneration. FASEB J. 2021;35:e21442. doi: 10.1096/fj.202002611R.
    1. Lou BS, Hsieh JH, Chen CM, Hou CW, Wu HY, Chou PY, et al. Helium/argon-generated cold atmospheric plasma facilitates cutaneous wound healing. Front Bioeng Biotechnol. 2020;8:683. doi: 10.3389/fbioe.2020.00683.
    1. Zhang M, Liu J, Cheng A, Deyoung SM, Chen X, Dold LH, et al. CAP interacts with cytoskeletal proteins and regulates adhesion-mediated ERK activation and motility. EMBO J. 2006;25:5284–93. doi: 10.1038/sj.emboj.7601406.
    1. Boeckmann L, Schäfer M, Bernhardt T, Semmler ML, Jung O, Ojak G, et al. Cold atmospheric pressure plasma in wound healing and cancer treatment. Appl Sci 2020;10:6898.
    1. Lee HR, Lee HY, Heo J, Jang JY, Shin YS, Kim CH. Liquid-type nonthermal atmospheric plasma enhanced regenerative potential of silk-fibrin composite gel in radiation-induced wound failure. Mater Sci Eng C Mater Biol Appl. 2021;128:112304. doi: 10.1016/j.msec.2021.112304.
    1. Kang SU, Kim HJ, Kim DH, Han CH, Lee YS, Kim C-H. Nonthermal plasma treated solution inhibits adipocyte differentiation and lipogenesis in 3T3-L1 preadipocytes via ER stress signal suppression. Sci Rep. 2018;8:2277. doi: 10.1038/s41598-018-20768-5.
    1. Won HR, Kang SU, Kim HJ, Jang JY, Shin YS, Kim CH. Non-thermal plasma treated solution with potential as a novel therapeutic agent for nasal mucosa regeneration. Sci Rep. 2018;8:13754. doi: 10.1038/s41598-018-32077-y.
    1. Won HR, Song EH, Won JE, Lee HY, Kang SU, Shin YS, et al. Liquid-type non-thermal atmospheric plasma ameliorates vocal fold scarring by modulating vocal fold fibroblast. Exp Biol Med. 2019;244:824–33. doi: 10.1177/1535370219850084.
    1. Yu Y, Tan M, Chen H, Wu Z, Xu L, Li J, et al. Non-thermal plasma suppresses bacterial colonization on skin wound and promotes wound healing in mice. J Huazhong Univ Sci Technol Med Sci = Hua zhong ke ji da xue xue bao Yi xue Ying De wen ban = Huazhong keji daxue xuebao Yixue Yingdewen ban. 2011;31:390–4. doi: 10.1007/s11596-011-0387-2.
    1. Lee HR, Lee YS, You YS, Huh JY, Kim K, Hong YC, et al. Antimicrobial effects of microwave plasma-activated water with skin protective effect for novel disinfectants in pandemic era. Sci Rep. 2022;12:5968. doi: 10.1038/s41598-022-10009-1.
    1. Kim S, Kim CH. Applications of plasma-activated liquid in the medical field. Biomedicines. 2021;9:1700.
    1. Zhou D, Shao L, Spitz DR. Reactive oxygen species in normal and tumor stem cells. Adv Cancer Res. 2014;122:1–67. doi: 10.1016/B978-0-12-420117-0.00001-3.
    1. Dunnill C, Patton T, Brennan J, Barrett J, Dryden M, Cooke J, et al. Reactive oxygen species (ROS) and wound healing: the functional role of ROS and emerging ROS-modulating technologies for augmentation of the healing process. Int Wound J. 2017;14:89–96. doi: 10.1111/iwj.12557.
    1. Balta E, Kramer J, Samstag Y. Redox regulation of the actin cytoskeleton in cell migration and adhesion: on the way to a spatiotemporal view. Front Cell Dev Biol. 2020;8:618261. doi: 10.3389/fcell.2020.618261.
    1. Antonucci S, Di Lisa F, Kaludercic N. Mitochondrial reactive oxygen species in physiology and disease. Cell Calcium. 2021;94:102344. doi: 10.1016/j.ceca.2020.102344.
    1. Cano Sanchez M, Lancel S, Boulanger E, Neviere R. Targeting Oxidative Stress and Mitochondrial Dysfunction in the Treatment of Impaired Wound Healing: A Systematic Review. Antioxidants (Basel) 2018;7:98. doi: 10.3390/antiox7080098.
    1. Andreyev AY, Kushnareva YE, Murphy AN, Starkov AA. Mitochondrial ROS Metabolism: 10 Years Later. Biochemistry (Mosc) 2015;80:517–31. doi: 10.1134/S0006297915050028.
    1. Sardella E, Mola MG, Gristina R, Piccione M, Veronico V, Bellis M, et al. A synergistic effect of reactive oxygen and reactive nitrogen species in plasma activated liquid media triggers astrocyte wound healing. Int J Mol Sci. 2020;21:3343.
    1. Stratmann B, Costea TC, Nolte C, Hiller J, Schmidt J, Reindel J, et al. Effect of cold atmospheric plasma therapy vs standard therapy placebo on wound healing in patients with diabetic foot ulcers: a randomized clinical trial. JAMA Netw Open. 2020;3:e2010411. doi: 10.1001/jamanetworkopen.2020.10411.
    1. Hong YC, Shin DH, Lee SJ, Kim YJ, Lee BJ, Uhm HS. Generation of high-power torch plasma by a 915 MHz microwave system. IEEE Trans Plasma Sci. 2011;39:1958–62. doi: 10.1109/TPS.2011.2162969.
    1. Kang SU, Kim DH, Lee YS, Huang M, Byeon HK, Lee S-H, et al. DIM-C-pPhtBu induces lysosomal dysfunction and unfolded protein response—mediated cell death via excessive mitophagy. Cancer Lett. 2021;504:23–36. doi: 10.1016/j.canlet.2021.01.005.
    1. Chang JW, Kang SU, Shin YS, Kim KI, Seo SJ, Yang SS, et al. Non-thermal atmospheric pressure plasma inhibits thyroid papillary cancer cell invasion via cytoskeletal modulation, altered MMP-2/-9/uPA activity. PLoS One. 2014;9:e92198. doi: 10.1371/journal.pone.0092198.

Source: PubMed

3
Subscribe