Next Article in Journal
Interleukin-10 Mitigates Doxorubicin-Induced Endoplasmic Reticulum Stress as Well as Cardiomyopathy
Next Article in Special Issue
Anti-Cancer Activity of the Combinational Treatment of Noozone Cold Plasma with p-FAK Antibody-Conjugated Gold Nanoparticles in OSCC Xenograft Mice
Previous Article in Journal
Ion Channels and Transporters as Therapeutic Agents: From Biomolecules to Supramolecular Medicinal Chemistry
Previous Article in Special Issue
New Approach against Chondrosoma Cells—Cold Plasma Treatment Inhibits Cell Motility and Metabolism, and Leads to Apoptosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Non-Invasive Physical Plasma Generated by a Medical Argon Plasma Device Induces the Expression of Regenerative Factors in Human Gingival Keratinocytes, Fibroblasts, and Tissue Biopsies

1
Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, 53111 Bonn, Germany
2
Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
3
Department of Orthodontics, University Hospital Bonn, 53111 Bonn, Germany
4
Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
5
Section of Experimental Dento-Maxillo-Facial Medicine, University Hospital Bonn, 53111 Bonn, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2022, 10(4), 889; https://doi.org/10.3390/biomedicines10040889
Submission received: 15 March 2022 / Revised: 7 April 2022 / Accepted: 11 April 2022 / Published: 13 April 2022
(This article belongs to the Special Issue The Advances of Cold Plasma in the Biomedicines)

Abstract

:
After oral surgery, intraoral wound healing and tissue regeneration is an important factor for the success of the entire therapy. In recent years, non-invasive medical plasma (NIPP) has been shown to accelerate wound healing, which would be particularly beneficial for patients with wound healing disorders. Since the application of NIPP in dentistry has not been sufficiently understood, the aim of the present study was to investigate the effect of a medical argon plasma device on gingival cells. Human gingival fibroblasts, keratinocytes, and tissue biopsies were treated with NIPP for different durations. Crucial markers associated with wound healing were examined at the mRNA and protein levels by real-time PCR, ELISA and immunohistochemistry. NIPP treatment led to an increase in Ki67 and MMP1 at mRNA and protein levels. NIPP application lasting longer than 60 s resulted in an increase in apoptotic genes at mRNA level and superficial damage to the epithelium in the tissue biopsies. Overall, our experimental setup demonstrated that NIPP application times of 30 s were most suitable for the treatment of gingival cells and tissue biopsies. Our study provides evidence for potential use of NIPP in dentistry, which would be a promising treatment option for oral surgery.

1. Introduction

In dentistry, especially in the field of oral and maxillofacial surgery, intraoral wound healing is a key factor for the success of an intervention. In particular, the regeneration of the soft tissue is essential for the underlying structures of the bone, as this is an important barrier against microorganisms, a large number of which reside in the oral cavity [1]. In addition, the gingiva is a barrier against antigens found in the air, which make their first contact with human organisms there before passing into the gastrointestinal and respiratory tracts [2].
The gingiva consists of a surface epithelium separated from the underlying dense connective tissue by a basal lamina [3]. The surface epithelium is a multilayered squamous epithelium that is keratinised in the masticatory areas and nonkeratinised in the other lining areas. For this reason, keratinocytes and fibroblasts are predominantly found in this tissue [3]. An important element of the connective tissue is collagen 1: Its main structural component collagen type 1 α1 (COL1A1) is produced by fibroblasts and can also be influenced by keratinocytes. [4,5].
The interaction of various factors contributes to wound healing: Various cytokines are involved not only in inflammatory responses, but also in critical functions of wound healing such as wound contraction or tissue remodeling [6,7,8,9,10]. During wound healing, the reorganization of collagen is essential for the entire tissue repair [11]. This process of remodeling is mainly controlled by matrix metalloproteinases (MMP)s, such as MMP1 [12]. The main characteristic of a healing wound is a high proliferation rate of dividing cells, as indicated by high levels of proliferation-associated proteins Ki67 and Proliferating Cell Nuclear Antigen (PCNA) [13,14]. Ki67 is found in all active cell cycle phases, but it is not present in quiescent cells [15]. PCNA is a nuclear protein that is obligatory for DNA synthesis and is mainly expressed during the longer G1 and S phases of the cell cycle [16].
Within the last 10 years, non-invasive physical plasma (NIPP) has become the focus of medical research. NIPP is a highly reactive, electrically conductive gas, also known as the 4th state of matter, which has various properties [17]. For example, antimicrobial, antioncogenic, and wound-healing effects have been described for the medical field [18,19,20,21,22,23]. Several types of NIPP devices have been designed: Plasma jets, Dielectric Barrier Discharges (DBD), and hybrid devices that combine both technologies [24]. In all devices, plasma is generated between the cathode and anode by a high-frequency voltage. In DBD devices, the treated object is the counter-electrode, whereas in plasma jets, a carrier gas is used to deliver NIPP from inside the device to the target [25]. Various clinical studies have also shown a supportive effect of different NIPP devices on gingival wound healing [26,27]. However, a more in-depth molecular investigation of the effect of NIPP on gingival cells and tissues has not yet been described. The aim of the study was therefore to investigate the effect of NIPP generated by a medical argon plasma device on human gingival keratinocytes (HGK) and human gingival fibroblasts (HGF) regarding crucial molecules involved in wound healing. Since apoptotic effects by NIPP have also been described [19], the effects on crucial apoptotic molecules, such as caspase (CASP)9 and CASP3, were also to be investigated in this study. In addition, the effect of NIPP should also be demonstrated histologically in tissue biopsies of gingival tissue to demonstrate the effect of a regular clinical use of NIPP in dentistry.

2. Materials and Methods

2.1. Cell Culture

HGF (HGF-1; CRL-2014) and HGK (Primary Gingival Keratinocytes; PCS-200-014) were obtained from ATCC (Manassas, VA, USA). HGF were propagated in Dulbecco’s modified essential medium (DMEM; Invitrogen, Waltham, MA, USA), containing 10% fetal bovine serum (FBS; Invitrogen, Waltham, MA, USA), and 1% penicillin/streptomycin (Invitrogen) at 37 °C with 5% CO2 and 95% humidity. The cultivation of HGK was performed under the same conditions in Keratinocyte Growth Medium 2 (PromoCell, Heidelberg, Germany), supplemented with 1% penicillin/streptomycin. The cell culture medium was changed every 2–3 days. For the experiments, 50,000 cells/well were each cultivated in 3.5 cm dishes (VWR, Radnor, PA, USA). One day prior to the experiments, FBS concentration of HGF was reduced to 1%.

2.2. NIPP Application

NIPP was generated by an argon plasma jet (kINPen med, neoplas med, Greifswald, Germany) at 4.0 L of argon per minute. The treatment of medium covered cells was performed at a distance of 2 cm (nozzle to cells), describing a spiral for the indicated times.

2.3. Analysis of mRNA Expression

One day after the experiments, the RNA was extracted using a RNeasy Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. An iScript™ Select cDNA Synthesis Kit (Bio-Rad Laboratories, Munich, Germany) was used to reverse transcribe 1 µg RNA into cDNA. A mixture of 12.5 µL SsoAdvanced™ Universal SYBR® Green Supermix (Bio-Rad), 2.5 µL commercially available primer (GAPDH, Ki67, PCNA, COL1A1, MMP1, CASP9, and CASP3; QuantiTect Primer Assay, Qiagen), and 9 µL deionised water were used to amplify 1 µL cDNA by the iCycler iQ™5 detection system (Bio-Rad). For real time PCR, the following protocol was used: 5 min heating at 95 °C, 40 cycles denaturation at 95 °C for 10 s and combined anealing/extension at 60 °C for 30 s. The data were evaluated using the comparative threshold cycle method.

2.4. Analysis of Ki67 and MMP1 Protein Levels

Ki67 protein levels were analysed in cell lysates using the Human Ki67 enzyme-linked immunoassay (ELISA) Kit (ab253221, abcam, Cambridge, UK). For quantification of MMP1 protein levels, cell supernatants were analysed using the Human Total MMP-1 DuoSet (ELISA) kit (DY901, Bio-Techne, Minneapolis, MI, USA) according to the manufacturer’s instructions. Absorbance was measured at 450 nm using a microplate reader (Epoch™ Microplate Spectrophotometer, BioTek Instruments, Winooski, VT, USA) and normalized to total protein concentration using Pierce BCA Protein Assay Kit (23227, Thermo Scientific, Pierce Biotechnology, Rockford, WA, USA). The absorbance was measured at 570 nm, as described above.

2.5. Caspase-3/7 Assay

Cells were seeded into 3.5 cm dishes, as described above, and were subsequently treated with NIPP. Immediately after treatment, cell suspension was transferred to 96 well plates and incubated for 24 h and 48 h. After incubation, medium was removed and 100 μL of Caspase 3/7 detection solution (CellEventTM Caspase 3/7 Green Detection Reagent, Thermo Fisher Scientific, Waltham, MA, USA) was added according to the manufacturer’s instructions for 45 min. Staurosporine (Sigma-Aldrich, St. Louis, MO, USA) treated cells served as positive control. Fluorescens was measured at 535 nm following excitation at 495 nm using a microplate reader (Tecan, Männedorf, Switzerland).

2.6. Staining of Tissue Biopsies

Human gingival biopsies, intraoperatively classified as tissue waste, were taken from five healthy patients during routine surgeries in the Department of Oral Surgery at Bonn University Hospital. The approval of the ethics committee of the University of Bonn (#111/17) and the written informed consent of the patients were obtained in advance. Tissue was rinsed with Phosphate buffered saline (PBS; Invitrogen) and cut into thirds. Two thirds were treated with NIPP using the spacer provided by the company; the other third served as a control group. The tissues were stored in 3.5 cm wells (VWR) in DMEM (Invitrogen), containing 1% FBS (Invitrogen), and 1% penicillin/streptomycin (Invitrogen) at 37 °C with 5% CO2 and 95% humidity for 24 h. Subsequently, the pieces of tissues were fixed in 4% paraformaldehyde (Sigma-Aldrich) for 2 d, hydrated, dehydrated in an ascending ethanol series (AppliChem, Darmstadt, Germany), and embedded in paraffin (McCormick Scientific, Richmond, IL, USA). The tissue was sliced into sections of 4 µm and mounted on glass slides (SuperFrost Plus; Thermo Fisher Scientific).
For histological analysis, sections were stained with hematoxylin and eosin (HE; Merck, Darmstadt, Germany), dehydrated, and mounted with DePeX (SERVA Electrophoresis GmbH, Heidelberg, Germany).
For immunohistochemistry, sections were deparaffinated, rehydrated, and rinsed with TBST (Merck) for 10 min. Afterwards, the endogenous peroxidase was blocked by using 0.3% methanol (AppliChem)/H2O2 (Merck) solution for 10 min. After another rinsing with TBST, the sections were blocked with 5% Bovine Serum Albumin Fraction V (BSA; Roche Diagnostics, Mannheim, Germany) in PBS for 30 min and incubated with primary antibodies rabbit anti-Ki67 (1:2000; abcam), or rabbit anti-MMP1 (1:600; abcam) in 1% BSA in a humid chamber at 4 °C overnight. Following this, the sections were rinsed with TBST and incubated with goat anti-rabbit IgG-HRP secondary antibody (Dako, Glostrup, Denmark) at room temperature for 30 min. The staining was visualized with 3.3′-diaminobenzidine chromogen (Thermo Fisher Scientific) and—after rinsing with TBST—counterstained with Mayer’s hematoxylin (Merck) for 5 s. After the rinsing and dehydration, the slides were mounted with DePeX. Analysis was performed using the Axioskop 2 microscope (Carl Zeiss, Jena, Germany) with an AxioCam MRc camera and the AxioVision 4.7 software (Carl Zeiss).

2.7. Statistical Analysis

For statistics, GraphPad Prism Software Version 7 (GraphPad Software, Inc., La Jolla, CA, USA) was used with one-way ANOVA and post hoc Dunnett’s multiple comparisons test. p values below 0.05 were defined as statistically significant.

3. Results

3.1. Analysis of Ki67 and PCNA

At first, we investigated the effect of NIPP on cellular growth. In HGF and HGK, treatment with NIPP caused an increase in Ki67 mRNA levels. In HGF, there was a dose-dependent increase of Ki67 mRNA for exposure times of 10 s to 30 s (Figure 1a). Longer treatment times resulted in a decrease in Ki67 mRNA regulation. In HGK, the dose-dependent mRNA upregulation was evident up to an application time of nearly 60 s (Figure 1b). The mRNA regulation of PCNA in HGF showed a similar pattern to Ki67 with a peak at 30 s (Figure 1c). Interestingly, in HGK, the PCNA expression maximum also occurred after 60 s of NIPP but was more pronounced than the Ki67 expression (Figure 1d).
Following the investigation of mRNA expression, we then focused on the examination of protein expression of Ki67.
In HGF protein expression of both one day and two days was similar to mRNA level at one day (Figure 2a,b). Interestingly, in contrast to mRNA data, 10 s of NIPP treatment showed a significant reduction in Ki67 protein expression after 48 h. However, the protein data confirmed that 30 s was the most effective NIPP treatment time in HGF.
In contrast, in HGK, Ki67 protein levels decreased in a dose-dependent manner after 24 h of incubation, indicating anti-proliferative effects of NIPP (Figure 2c). Interestingly, Ki67 protein expression after 48 h correlated with mRNA data from the 24 h incubation (Figure 1b), demonstrating a dose-dependent stimulatory effect with a maximum at 30 s NIPP treatment (Figure 2d). Similar to the 24 h incubation data in HGK, 90 s NIPP led to a strong reduction of Ki67 protein levels by almost 50% at 48 h (Figure 2d).

3.2. Analysis of COL1A1 and MMP1

Next, we focused on the study of marker molecules related to tissue remodeling. First, we examined the mRNA expression of COL1A1 as a major component of gingival connective tissue. Interestingly, NIPP had little effect on mRNA expression in HGF after 24 h (Figure 3a). In HGK, 10 s of NIPP caused a reduction in COL1A1 mRNA expression after 24 h—however, only very limited effects of NIPP were observed in HGK (Figure 3b).
In addition, we investigated the influence of NIPP on MMP1, which plays a pivotal role in collagen degradation and remodeling.
In HGF, there was a significant increase in MMP1 regulation in relation to increasing NIPP treatment time (Figure 3c). In HGK, a similar effect was seen, but an increase in MMP1 mRNA regulation was only seen from 30 s NIPP treatment after 24 h (Figure 3d).
The NIPP effect on the modulation of MMP1 expression was also detected at the protein level.
In HGF, the dose-dependent effect of the NIPP application time was already apparent after 24 h (Figure 4a) and was also still visible after two days, with a peak at 90 s of NIPP (Figure 4b). HGK showed a similar pattern, with 90 s NIPP also having the strongest effect on MMP1 protein levels at one and two days (Figure 4c,d).

3.3. Examination of CASP9 and CASP3

Finally, we investigated the influence of NIPP on apoptotic genes, as NIPP is also known for its apoptotic effect in cancer cells. It was shown for application times of 90 s NIPP in HGF and 60 s NIPP, as well as 90 s NIPP in HGK, that there was an upregulation in the mRNA expression of CASP9 (Figure 5a,b). Interestingly, this effect could not be detected with HGF in the mRNA expression of CASP3, which is regulating the end of the apoptotic cascade (Figure 5c). In comparison, CASP3 mRNA was upregulated in HGK at application times of 60 s. Interestingly, the effect of 90 s NIPP was significantly lower on CASP3 expression (Figure 5d).
The apoptotic effect of NIPP on the cells was measured with a caspase-3/7 assay. Surprisingly, the slight apoptotic effect of NIPP on the cells could not be confirmed: Both HGF and HGK showed no significant increase of active caspase 3/7 after 24 h and 48 h compared to the untreated control (Figure 6a–d).

3.4. Examination of Tissue Biopsies

To evaluate the in vitro data, gingival tissue was treated with NIPP in vitro and analysed. For this purpose, tissue biopsies were treated with NIPP immediately after collection, incubated for 24 h, and examined histologically. Analysis of the sections showed that an application of 60 s NIPP led to damage of the superficial keratinocyte layer and slight dissolution of the suprabasal layers in the tissue (Figure 7).
The expression of Ki67 could also be detected in the tissue biopsies. Since the tissues were stored in cell culture medium, the control group also showed Ki67 expression. However, 30 s NIPP application, in particular, caused an increase in Ki67 expression in the fibroblasts of the connective tissue and the basal layer of the keratinocytes. A 60 s NIPP application resulted in increased immunostaining of the superficial keratinocyte layers and reduced staining in basal layers. (Figure 8).
Finally, we also investigated the expression of MMP1 in tissue biopsies. Here, in particular, the strong dose-dependent effects of NIPP could be demonstrated, which were especially evident in the subepithelial layer. Additionally, some keratinocytes were stained in the basal layer of epithelium. A 30 s NIPP treatment resulted in an increase in MMP1 staining throughout the whole lamina propria, and this was even more pronounced with a 60 s treatment (Figure 9).

4. Discussion

In the present study we have shown the proliferation-promoting effect of NIPP generated by an argon plasma device on gingival tissue concerning crucial markers regarding tissue regeneration and wound healing (Figure 10). In previous in vitro studies, we have shown similar effects of DBD-generated NIPP on hard tissue cells [28,29]. However, as the soft tissue is crucial for the healing of the underlying hard tissue without complications, the aim of this study was to investigate the effect on gingival cells and tissue.
First, we focused on mRNA expression of different molecules crucial for wound healing. For this reason, we examined the mRNA expression of Ki67 and PCNA, which are well-established markers for the detection of proliferating cells [16] and additionally focused on Ki67 protein expression. In HGF cells, the proliferative effect is strongest with 30 s NIPP treatment and decreases with longer application times. In HGK, similar effects were observed, but Ki67 protein expression showed the same pattern as mRNA expression only after 48 h, which might be due to the time needed to transcribe the mRNA information into the protein.
The proliferation-promoting effect of NIPP has already been described in the literature [29,30,31,32,33,34,35,36]. In previous in vitro studies, we have shown an upregulation of Ki67 and PCNA expression after 60 s of NIPP treatment at 24 h in PDL cells, gingival keratinocytes, and cementoblasts using a DBD [30,35,37]. Interestingly, after one day in HGK using a DBD, 60 s NIPP treatment revealed similar results in regard to Ki67 mRNA expression, as we have shown in the present study, using an argon plasma jet. However, mRNA expression of PCNA after 60 s of NIPP treatment was higher using a plasma jet than using a DBD [37]. Further studies to systematically compare the two devices are needed to better understand the exact effect of NIPP on gingival cells. Since in the present study we demonstrated an effect on Ki67 and PCNA for only 10 s of NIPP treatment after one day, even short applications seem to have an effect, which would facilitate clinical use. Other authors have shown this proliferative effect with as little as 3 s of NIPP treatment in vivo after 6 days, using the same device as we have used [32]. But different applications of NIPP have also been described in the literature: Park et al. (2019) showed an upregulation of PCNA protein expression in adipose tissue-derived stem cells after NIPP treatment of 50 s per h for 10 times at 24 h and 72 h, respectively [38]. For this reason, further studies are necessary to investigate different application times, such as 20 s, 40 s, and 50 s of NIPP treatment. It is also possible that the maximum of mRNA and protein expression is not at a treatment time of 30 s, but between 10 s and 30 s or 30 s and 60 s. Only further studies can illuminate this. In addition, the different effects of the various devices on the same cells should also be systematically examined.
It must also be considered that protein expression must also have an effect on cell number. However, in the previous in vitro studies mentioned above, we were also able to show that the NIPP-regulated upregulation of Ki67 and PCNA correlates with an increasing cell number: Thus, a significant increase in cell number was shown after 24 h in osteoblast-like cells [29]. However, a systematic investigation of the cell number with regard to different time points should also be the target of further studies with HGF and HGK.
Furthermore, the influence of NIPP on other components of in vitro cell regeneration, such as the metabolic activity of the cells or cell migration, has been described in the literature: Cui et al. have described increased cell migration and higher levels of angiogenic growth factors in human keratinocytes treated with LTP for 30 s, 60 s and 180 s using a DBD plasma device [36]. Liu et al. used MTT to demonstrate an increase in cell viability in mouse fibroblasts in vitro after 48 h when exposed to an argon plasma jet at 0.5 L/min from 0–30 s: They showed a peak at 15 s, which might be due to the different device or a different, not specified distance to the cells [39]. Nevertheless, it must be taken into account that different cell types and cells from different tissues can show different cell responses to the same stimuli—as already shown for HGF and HGK.
Further on, we focused on COL1A1 and MMP1, which are both important regulators of early wound healing [40,41]. COL1A1 is mainly produced by fibroblasts rather than keratinocytes, although keratinocytes are able to control the production of COL1A1 in fibroblasts [4]. However, we decided to also investigate the mRNA expression of COL1A1 in keratinocytes, as platelet-released growth factors (e.g., platelet-rich fibrin) have been described to stimulate COL1A1 mRNA expression in keratinocytes [42]. Interestingly, COL1A1 mRNA regulation was not substantially affected by plasma in either HGF or HGK. In previous in vitro studies, we found an upregulation of COL1A1 mRNA after NIPP treatment of osteoblasts, PDL cells, and cementoblasts using a DBD [29,30,35]. In addition, dermal and gingival fibroblasts have both been described to show increased mRNA expression of COL1A1 after NIPP application for 1, 3 and 5 min, and 1, 2, and 4 min in vitro [43,44]. We can only speculate about the reasons for the differences in the literature, but on the one hand cells seem to react differently to NIPP influence, while on the other hand all the authors have used different NIPP devices and different experimental settings: Osteoblasts, PDL cells and cementoblasts were stimulated by a DBD [29,30,35]; dermal fibroblasts were stimulated by a non-commercial microwave plasma generator [43], and HGF were stimulated by an experimental plasma jet supplied by compressed air at 1 L/min [44]. Since our device operates with argon gas at higher gas flow (4 L/min), additional research is needed to further decipher the differences of the various devices on gingival cells.
Furthermore, MMP1 was upregulated by NIPP both at the mRNA level and at the protein level in the cell culture supernatant in a dose-dependent manner. We have already been able to demonstrate the promoting effect of NIPP on MMP1 in cells of the periodontal ligament and in hard tissue cells using a DBD in vitro [29,30]. Therefore, we can assume that the effect of NIPP on the regulation of MMP1 is independent of the technology used to generate NIPP. However, as elevated MMP1 levels have also been associated with periodontitis [45], it must be ruled out that NIPP is not harmful to gingival cells and tissue. However, since we have shown that short treatment times lead to early MMP1 upregulation in HGK and HGF without affecting the regulation of apoptotic markers, this seems to be ruled out. Other methods that promote wound healing also have an effect on increased MMP1 secretion, such as the use of platelet-rich plasma [46].
Longer NIPP applications, such as 60 s and especially 90 s treatment, are associated with upregulation of CASP3 and CASP9 mRNA. Interestingly, this does not result in higher caspase-3/7 activity. It is possible that NIPP seems to irritate the cells but prevents the cells from actually becoming apoptotic. This underlines the non-invasive effect of NIPP.
Apoptotic effects of NIPP have been abundantly described in the literature, especially for malignant cells [47,48,49]. In non-malignant cells, the antiproliferative effect is evident when NIPP is applied for a long time: In HaCaT keratinocytes, 180 s NIPP treatments have been described as apoptotic after 24 h [50]. Additionally, Weiss et al. have shown a significant decrease in the cell number of fibroblasts after 72 and 120 h when treated for 120 s [51]. Further studies are needed here to determine whether the longer NIPP treatment in HGK and HGF is also reflected in a decrease in cell number. It is possible that the direct NIPP effects regarding apoptosis might already be detectable a few hours after NIPP application. However, in previous studies, we also observed downregulation of mRNA regulation of apoptotic genes and an anti-apoptotic effect in PDL cells and MG63 cells by NIPP [29,30]. However, one reason for the difference could be the device—in those studies an ambient air-controlled DBD was used, whereas in the present study we have used an argon plasma jet. Weiss et al. and Schmidt at al. have also used plasma jets [50,51]. Interestingly, in the literature overall harmful effects of NIPP jets on gingival cells have been described: Lee et al. demonstrate a harmful effect of compressed-air plasma (3 L/min) on HGF viability in vitro [52]. However, in this study, in contrast to our study design, the cells were not covered with medium during the NIPP treatment. Regarding our results, it must also be borne in mind that distance and continuous movement play a role. Thus, we were also able to induce apoptotic effects on HGF and HGK with our NIPP device at closer distances without continuous movement of the jet (data not shown). In addition, it must also be taken into account that the gas flow of 4 L/min might dry out the medium, even though we did not observe any significant difference in the amount of medium after NIPP treatment compared to the untreated control when collecting the cell culture supernatants for the ELISA experiments. However, further in vitro studies between medium-covered and non-covered cells are necessary to fully unravel the effects of gas flow and NIPP on the cells. Additionally, as there is no gas flow with DBD devices, a systematic investigation of the influence of these devices on gingival cells would help to further unravel the effect of NIPP in dentistry.
In the present study, we also focused on the examination of tissue biopsies where the monolayer results could be confirmed. Interestingly, we observed a slight injury of the superficial keratinocyte layer after 60 s NIPP, which could be an effect of the direct treatment. The difference from our in vitro experiments could be due to the fact that the cells on the surface already become apoptotic immediately after treatment or that the damage is attributed to a different mechanism. Further research on apoptosis is necessary to better understand this difference. Jablonowski et al. have, however, described these side effects of NIPP generated by a medical argon plasma device in an in vivo study: 10 s and 60 s NIPP cause focal mucosal erosion with superficial ulceration after 1 d in mice, but heal within a week without complications [53]. Furthermore, this superficial lesion was considered non-carcinogenic [54]. It seems that these superficial injuries do not contradict the clinical use of NIPP: Clinical studies have shown wound-healing effects on gingival wound healing regarding application times of 60 s using a plasma jet and 120 s using a DBD [26,27]. Since clinical healing of the gingiva was observed even with prolonged applications of NIPP, the observed superficial lesions do not seem to be relevant. This is confirmed by our results showing expression of Ki67 and MMP1 protein in the tissue layers beneath the epithelium. These results correspond to the results of other authors who have found NIPP effects at tissue depths of up to 2 mm [55]. However, further in vivo and in vitro studies are necessary to better understand the direct effect of NIPP on gingival wound healing.
Nevertheless, other methods of applying NIPP would be possible in dentistry, which might achieve a similar effect: Lou et al. observed an increase in Ki67 protein expression after 24 h using NIPP activated medium [33]. This possibility of applying multibiological functions to the fluid through NIPP treatment would greatly simplify its application in dentistry, allowing it to be used as a mouth rinse [56].
Whatever the method of applying NIPP, the use of NIPP is a cost-effective and time-saving method to improve gingival wound healing. In particular, patients with impaired wound healing, such as diabetics or oncological patients, could benefit. A reduction of these wound-healing disorders would not only have a psychological benefit for the patients but would also help to save costs in the health care system [57,58].
However, this study has some limitations: For protein analysis, we focused only on the selected markers Ki67 and MMP1. Since PCNA mRNA was also strongly regulated by NIPP, the analysis of protein levels after NIPP treatment would also have been useful. However, we followed the publication of Bologna-Molina et al., who identified both markers as similar proliferation markers, but gave Ki67 a higher specificity regarding ameloblastic cells [16]. Further studies comparing Ki67 and PCNA directly in HGF and HGK could be interesting to further decipher the NIPP effect on cell proliferation in gingival cells. Additionally, we used tissue biopsies to study NIPP effects. Since this tissue is only nourished by the cell culture medium, only marginal conclusions can be drawn about the biological effect within a living organism. This had an impact on the Ki67 staining, as the cells in the tissue were constantly proliferating due to storage in medium, requiring us to adjust the antibody dilution to visualise differences between the groups. For this reason, fibroblast staining within the tissue is low. However, the penetration depth of NIPP has already been demonstrated in the literature [59]. Additionally, we studied tissue biopsies at one day because of the risk of tissue degradation in the humified incubator, regarding a longer treatment time. For this reason, further in vivo experiments with a focus on intraoral wound healing are necessary to further understand the effect of NIPP. However, the aim of the present study was to use human samples, which is more representative of the patients who will be treated with NIPP. Furthermore, it must be borne in mind that commercially available cells were used for the monolayer experiments. Although we were able to confirm cell culture results in the tissue model with regard to MMP1 and Ki67, primary HGF and primary HGK may show different results of gene and protein expression. Further studies should therefore investigate this point in more detail.

5. Conclusions

Within the limits of our in vitro study, we could demonstrate that NIPP generated by a medical argon plasma device promotes wound healing-associated processes in human gingival cells and tissue. Although the exact effects of different NIPP devices on the gingiva are not yet known, it seems that argon-based plasma jets offer promising prospects for patients suffering from delayed wound healing and, last but not least, offer all patients the prospect of not being limited too long after surgical interventions.

Author Contributions

Conceptualization, B.E., F.-J.K. and M.N.; Methodology, B.E., J.M., M.B.S., F.-J.K. and M.N.; software, B.E.; Validation, B.E., M.B.S., W.G., J.D., F.-J.K. and M.N.; Formal Analysis, B.E., M.B.S., W.G., A.M., J.D., F.-J.K. and M.N.; Investigation, B.E. and M.N.; Resources, B.E., F.-J.K. and M.N.; Data Curation, B.E., J.M., J.D., W.G., M.B.S., A.M., F.-J.K. and M.N.; Writing—Original Draft Preparation, B.E., J.M., M.B.S., W.G., A.M., J.D., F.-J.K. and M.N.; Writing—Review and Editing, B.E., J.M., M.B.S., W.G., A.M., J.D., F.-J.K. and M.N.; Visualization, B.E., J.D., M.B.S., F.-J.K. and M.N.; Supervision, B.E.; Project Administration, F.-J.K. and M.N.; funding acquisition, B.E. All authors have read and agreed to the published version of the manuscript.

Funding

B.E. was supported by local funding (BONFOR, O-133.0001).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the Ethics Committee of the University Hospital of Bonn (#111/17).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Inka Bay-Müller and Rachana Subedi for the valuable technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Methé, B.A.; Nelson, K.E.; Pop, M.; Creasy, H.H.; Giglio, M.G.; Huttenhower, C.; Gevers, D.; Petrosino, J.F.; Abubucker, S.; Badger, J.H.; et al. A Framework for Human Microbiome Research. Nature 2012, 486, 215–221. [Google Scholar] [CrossRef] [Green Version]
  2. Dutzan, N.; Konkel, J.E.; Greenwell-Wild, T.; Moutsopoulos, N.M. Characterization of the Human Immune Cell Network at the Gingival Barrier. Mucosal Immunol. 2016, 9, 1163–1172. [Google Scholar] [CrossRef]
  3. Winning, T.A.; Townsend, G.C. Oral Mucosal Embryology and Histology. Clin. Dermatol. 2000, 18, 499–511. [Google Scholar] [CrossRef]
  4. Russo, B.; Brembilla, N.C.; Chizzolini, C. Interplay Between Keratinocytes and Fibroblasts: A Systematic Review Providing a New Angle for Understanding Skin Fibrotic Disorders. Front. Immunol. 2020, 11, 648. [Google Scholar] [CrossRef]
  5. Diegelmann, R.F.; Cohen, I.K.; McCoy, B.J. Growth Kinetics and Collagen Synthesis of Normal Skin, Normal Scar and Keloid Fibroblasts in Vitro. J. Cell. Physiol. 1979, 98, 341–346. [Google Scholar] [CrossRef]
  6. Hübner, G.; Brauchle, M.; Smola, H.; Madlener, M.; Fässler, R.; Werner, S. Differential Regulation of Pro-Inflammatory Cytokines during Wound Healing in Normal and Glucocorticoid-Treated Mice. Cytokine 1996, 8, 548–556. [Google Scholar] [CrossRef]
  7. McFarland-Mancini, M.M.; Funk, H.M.; Paluch, A.M.; Zhou, M.; Giridhar, P.V.; Mercer, C.A.; Kozma, S.C.; Drew, A.F. Differences in Wound Healing in Mice with Deficiency of IL-6 versus IL-6 Receptor. J. Immunol. 2010, 184, 7219–7228. [Google Scholar] [CrossRef] [Green Version]
  8. Takamiya, M.; Fujita, S.; Saigusa, K.; Aoki, Y. Simultaneous Detection of Eight Cytokines in Human Dermal Wounds with a Multiplex Bead-Based Immunoassay for Wound Age Estimation. Int. J. Legal Med. 2008, 122, 143–148. [Google Scholar] [CrossRef]
  9. Low, Q.E.H.; Drugea, I.A.; Duffner, L.A.; Quinn, D.G.; Cook, D.N.; Rollins, B.J.; Kovacs, E.J.; DiPietro, L.A. Wound Healing in MIP-1α−/− and MCP-1−/− Mice. Am. J. Pathol. 2001, 159, 457–463. [Google Scholar] [CrossRef]
  10. Futagami, A.; Ishizaki, M.; Fukuda, Y.; Kawana, S.; Yamanaka, N. Wound Healing Involves Induction of Cyclooxygenase-2 Expression in Rat Skin. Lab. Investig. 2002, 82, 1503–1513. [Google Scholar] [CrossRef] [Green Version]
  11. Beare, A.H.M.; O’Kane, S.; Krane, S.M.; Ferguson, M.W.J. Severely Impaired Wound Healing in the Collagenase-Resistant Mouse. J. Investig. Dermatol. 2003, 120, 153–163. [Google Scholar] [CrossRef] [PubMed]
  12. Saffarian, S.; Collier, I.E.; Marmer, B.L.; Elson, E.L.; Goldberg, G. Interstitial Collagenase Is a Brownian Ratchet Driven by Proteolysis of Collagen. Science 2004, 306, 108–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Schlüter, C.; Duchrow, M.; Wohlenberg, C.; Becker, M.H.; Key, G.; Flad, H.D.; Gerdes, J. The Cell Proliferation-Associated Antigen of Antibody Ki-67: A Very Large, Ubiquitous Nuclear Protein with Numerous Repeated Elements, Representing a New Kind of Cell Cycle-Maintaining Proteins. J. Cell Biol. 1993, 123, 513–522. [Google Scholar] [CrossRef]
  14. Hergott, G.J.; Kalnins, V.I. Expression of Proliferating Cell Nuclear Antigen in Migrating Retinal Pigment Epithelial Cells during Wound Healing in Organ Culture. Exp. Cell Res. 1991, 195, 307–314. [Google Scholar] [CrossRef]
  15. Scholzen, T.; Gerdes, J. The Ki-67 Protein: From the Known and the Unknown. J. Cell. Physiol. 2000, 182, 311–322. [Google Scholar] [CrossRef]
  16. Bologna-Molina, R.; Mosqueda-Taylor, A.; Molina-Frechero, N.; Mori-Estevez, A.D.; Sánchez-Acuña, G. Comparison of the Value of PCNA and Ki-67 as Markers of Cell Proliferation in Ameloblastic Tumor. Med. Oral Patol. Oral Cir. Bucal 2013, 18, e174–e179. [Google Scholar] [CrossRef] [PubMed]
  17. Braný, D.; Dvorská, D.; Halašová, E.; Škovierová, H. Cold Atmospheric Plasma: A Powerful Tool for Modern Medicine. Int. J. Mol. Sci. 2020, 21, 2932. [Google Scholar] [CrossRef] [Green Version]
  18. Kniha, K.; Möhlhenrich, S.C.; Bock, A.; Ayoub, N.; Modabber, A.; Hölzle, F.; Conrads, G.; Goloborodko, E. Evaluation of the Bactericidal Effect of Cold Atmospheric Pressure Plasma on Contaminated Human Bone: An in Vitro Study. Br. J. Oral Maxillofac. Surg. 2020, 58, 329–333. [Google Scholar] [CrossRef]
  19. Ma, Y.; Ha, C.S.; Hwang, S.W.; Lee, H.J.; Kim, G.C.; Lee, K.-W.; Song, K. Non-Thermal Atmospheric Pressure Plasma Preferentially Induces Apoptosis in P53-Mutated Cancer Cells by Activating ROS Stress-Response Pathways. PLoS ONE 2014, 9, e91947. [Google Scholar] [CrossRef]
  20. Stratmann, B.; Costea, T.-C.; Nolte, C.; Hiller, J.; Schmidt, J.; Reindel, J.; Masur, K.; Motz, W.; Timm, J.; Kerner, W.; 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. [Google Scholar] [CrossRef]
  21. Weiss, M.; Daeschlein, G.; Kramer, A.; Burchardt, M.; Brucker, S.; Wallwiener, D.; Stope, M.B. Virucide Properties of Cold Atmospheric Plasma for Future Clinical Applications. J. Med. Virol. 2017, 89, 952–959. [Google Scholar] [CrossRef] [PubMed]
  22. Haralambiev, L.; Neuffer, O.; Nitsch, A.; Kross, N.C.; Bekeschus, S.; Hinz, P.; Mustea, A.; Ekkernkamp, A.; Gümbel, D.; Stope, M.B. Inhibition of Angiogenesis by Treatment with Cold Atmospheric Plasma as a Promising Therapeutic Approach in Oncology. Int. J. Mol. Sci. 2020, 21, 7098. [Google Scholar] [CrossRef] [PubMed]
  23. Haralambiev, L.; Wien, L.; Gelbrich, N.; Kramer, A.; Mustea, A.; Burchardt, M.; Ekkernkamp, A.; Stope, M.B.; Gümbel, D. Effects of Cold Atmospheric Plasma on the Expression of Chemokines, Growth Factors, TNF Superfamily Members, Interleukins, and Cytokines in Human Osteosarcoma Cells. Anticancer Res. 2019, 39, 151–157. [Google Scholar] [CrossRef] [PubMed]
  24. Gan, L.; Zhang, S.; Poorun, D.; Liu, D.; Lu, X.; He, M.; Duan, X.; Chen, H. Medical Applications of Nonthermal Atmospheric Pressure Plasma in Dermatology. JDDG J. Dtsch. Dermatol. Ges. 2018, 16, 7–13. [Google Scholar] [CrossRef] [Green Version]
  25. Heinlin, J.; Isbary, G.; Stolz, W.; Morfill, G.; Landthaler, M.; Shimizu, T.; Steffes, B.; Nosenko, T.; Zimmermann, J.; Karrer, S. Plasma Applications in Medicine with a Special Focus on Dermatology. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 1–11. [Google Scholar] [CrossRef]
  26. Pekbağrıyanık, T.; Dadas, F.K.; Enhoş, Ş. Effects of Non-Thermal Atmospheric Pressure Plasma on Palatal Wound Healing of Free Gingival Grafts: A Randomized Controlled Clinical Trial. Clin. Oral Investig. 2021, 25, 6269–6278. [Google Scholar] [CrossRef]
  27. Kusakci-Seker, B.; Demirayak-Akdemir, M. The Effect of Non-Thermal Atmospheric Pressure Plasma Application on Wound Healing after Gingivectomy. Int. Wound J. 2020, 17, 1376–1383. [Google Scholar] [CrossRef]
  28. Eggers, B.; Wagenheim, A.-M.; Jung, S.; Kleinheinz, J.; Nokhbehsaim, M.; Kramer, F.-J.; Sielker, S. Effect of Cold Atmospheric Plasma (CAP) on Osteogenic Differentiation Potential of Human Osteoblasts. Int. J. Mol. Sci. 2022, 23, 2503. [Google Scholar] [CrossRef]
  29. Eggers, B.; Marciniak, J.; Memmert, S.; Kramer, F.J.; Deschner, J.; Nokhbehsaim, M. The Beneficial Effect of Cold Atmospheric Plasma on Parameters of Molecules and Cell Function Involved in Wound Healing in Human Osteoblast-like Cells in Vitro. Odontology 2020, 108, 607–616. [Google Scholar] [CrossRef] [Green Version]
  30. Kleineidam, B.; Nokhbehsaim, M.; Deschner, J.; Wahl, G. Effect of Cold Plasma on Periodontal Wound Healing-an in Vitro Study. Clin. Oral Investig. 2019, 23, 1941–1950. [Google Scholar] [CrossRef]
  31. Delben, J.A.; Zago, C.E.; Tyhovych, N.; Duarte, S.; Vergani, C.E. Effect of Atmospheric-Pressure Cold Plasma on Pathogenic Oral Biofilms and In Vitro Reconstituted Oral Epithelium. PLoS ONE 2016, 11, e0155427. [Google Scholar] [CrossRef]
  32. Schmidt, A.; von Woedtke, T.; Vollmar, B.; Hasse, S.; Bekeschus, S. Nrf2 Signaling and Inflammation Are Key Events in Physical Plasma-Spurred Wound Healing. Theranostics 2019, 9, 1066–1084. [Google Scholar] [CrossRef] [PubMed]
  33. Lou, B.-S.; Hsieh, J.-H.; Chen, C.-M.; Hou, C.-W.; Wu, H.-Y.; Chou, P.-Y.; Lai, C.-H.; Lee, J.-W. Helium/Argon-Generated Cold Atmospheric Plasma Facilitates Cutaneous Wound Healing. Front. Bioeng. Biotechnol. 2020, 8, 683. [Google Scholar] [CrossRef] [PubMed]
  34. Shao, P.-L.; Liao, J.-D.; Wu, S.-C.; Chen, Y.-H.; Wong, T.-W. Microplasma Treatment versus Negative Pressure Therapy for Promoting Wound Healing in Diabetic Mice. Int. J. Mol. Sci. 2021, 22, 10266. [Google Scholar] [CrossRef] [PubMed]
  35. Eggers, B.; Marciniak, J.; Deschner, J.; Stope, M.B.; Mustea, A.; Kramer, F.-J.; Nokhbehsaim, M. Cold Atmospheric Plasma Promotes Regeneration-Associated Cell Functions of Murine Cementoblasts In Vitro. Int. J. Mol. Sci. 2021, 22, 5280. [Google Scholar] [CrossRef] [PubMed]
  36. Cui, H.S.; Cho, Y.S.; Joo, S.Y.; Mun, C.H.; Seo, C.H.; Kim, J.-B. Wound Healing Potential of Low Temperature Plasma in Human Primary Epidermal Keratinocytes. Tissue Eng. Regen. Med. 2019, 16, 585–593. [Google Scholar] [CrossRef] [PubMed]
  37. Eggers, B.; Stope, M.B.; Mustea, A.; Nokhbehsaim, M.; Heim, N.; Kramer, F.-J. Non-Invasive Physical Plasma Treatment after Tooth Extraction in a Patient on Antiresorptive Medication Promotes Tissue Regeneration. Appl. Sci. 2022, 12, 3490. [Google Scholar] [CrossRef]
  38. Park, J.; Lee, H.; Lee, H.J.; Kim, G.C.; Kim, S.-S.; Han, S.; Song, K. Non-Thermal Atmospheric Pressure Plasma Is an Excellent Tool to Activate Proliferation in Various Mesoderm-Derived Human Adult Stem Cells. Free Radic. Biol. Med. 2019, 134, 374–384. [Google Scholar] [CrossRef]
  39. Liu, J.-R.; Xu, G.-M.; Shi, X.-M.; Zhang, G.-J. Low Temperature Plasma Promoting Fibroblast Proliferation by Activating the NF-ΚB Pathway and Increasing CyclinD1 Expression. Sci. Rep. 2017, 7, 11698. [Google Scholar] [CrossRef] [Green Version]
  40. Soo, C.; Shaw, W.W.; Zhang, X.; Longaker, M.T.; Howard, E.W.; Ting, K. Differential Expression of Matrix Metalloproteinases and Their Tissue-Derived Inhibitors in Cutaneous Wound Repair. Plast. Reconstr. Surg. 2000, 105, 638–647. [Google Scholar] [CrossRef]
  41. Nuytinck, L.; Freund, M.; Lagae, L.; Pierard, G.E.; Hermanns-Le, T.; De Paepe, A. Classical Ehlers-Danlos Syndrome Caused by a Mutation in Type I Collagen. Am. J. Hum. Genet. 2000, 66, 1398–1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Bayer, A.; Wijaya, B.; Möbus, L.; Rademacher, F.; Rodewald, M.; Tohidnezhad, M.; Pufe, T.; Drücke, D.; Gläser, R.; Harder, J. Platelet-Released Growth Factors and Platelet-Rich Fibrin Induce Expression of Factors Involved in Extracellular Matrix Organization in Human Keratinocytes. Int. J. Mol. Sci. 2020, 21, 4404. [Google Scholar] [CrossRef]
  43. Kim, H.-Y.; Agrahari, G.; Lee, M.J.; Tak, L.-J.; Ham, W.-K.; Kim, T.-Y. Low-Temperature Argon Plasma Regulates Skin Moisturizing and Melanogenesis-Regulating Markers through Yes-Associated Protein. Int. J. Mol. Sci. 2021, 22, 1895. [Google Scholar] [CrossRef] [PubMed]
  44. Kwon, J.-S.; Kim, Y.H.; Choi, E.H.; Kim, C.-K.; Kim, K.-N.; Kim, K.-M. Non-Thermal Atmospheric Pressure Plasma Increased MRNA Expression of Growth Factors in Human Gingival Fibroblasts. Clin. Oral Investig. 2016, 20, 1801–1808. [Google Scholar] [CrossRef] [PubMed]
  45. Nokhbehsaim, M.; Nogueira, A.V.B.; Damanaki, A.; Dalagiorgou, G.; Eick, S.; Adamopoulos, C.; Piperi, C.; Basdra, E.K.; Papavassiliou, A.G.; Deschner, J. Regulation of Matrix Metalloproteinase-1 by Filifactor Alocis in Human Gingival and Monocytic Cells. Clin. Oral Investig. 2020, 24, 1987–1995. [Google Scholar] [CrossRef] [PubMed]
  46. Cho, E.B.; Park, G.S.; Park, S.S.; Jang, Y.J.; Kim, K.H.; Kim, K.J.; Park, E.J. Effect of Platelet-Rich Plasma on Proliferation and Migration in Human Dermal Fibroblasts. J. Cosmet. Dermatol. 2019, 18, 1105–1112. [Google Scholar] [CrossRef]
  47. Haralambiev, L.; Wien, L.; Gelbrich, N.; Lange, J.; Bakir, S.; Kramer, A.; Burchardt, M.; Ekkernkamp, A.; Gümbel, D.; Stope, M.B. Cold Atmospheric Plasma Inhibits the Growth of Osteosarcoma Cells by Inducing Apoptosis, Independent of the Device Used. Oncol. Lett. 2020, 19, 283–290. [Google Scholar] [CrossRef] [Green Version]
  48. Almeida-Ferreira, C.; Silva-Teixeira, R.; Gonçalves, A.C.; Marto, C.M.; Sarmento-Ribeiro, A.B.; Caramelo, F.; Botelho, M.F.; Laranjo, M. Cold Atmospheric Plasma Apoptotic and Oxidative Effects on MCF7 and HCC1806 Human Breast Cancer Cells. Int. J. Mol. Sci. 2022, 23, 1698. [Google Scholar] [CrossRef]
  49. Gümbel, D.; Suchy, B.; Wien, L.; Gelbrich, N.; Napp, M.; Kramer, A.; Ekkernkamp, A.; Daeschlein, G.; Stope, M.B. Comparison of Cold Atmospheric Plasma Devices’ Efficacy on Osteosarcoma and Fibroblastic In Vitro Cell Models. Anticancer Res. 2017, 37, 5407–5414. [Google Scholar]
  50. Schmidt, A.; Bekeschus, S.; Jarick, K.; Hasse, S.; von Woedtke, T.; Wende, K. Cold Physical Plasma Modulates P53 and Mitogen-Activated Protein Kinase Signaling in Keratinocytes. Oxid. Med. Cell. Longev. 2019, 2019, 7017363. [Google Scholar] [CrossRef] [Green Version]
  51. Weiss, M.; Barz, J.; Ackermann, M.; Utz, R.; Ghoul, A.; Weltmann, K.-D.; Stope, M.B.; Wallwiener, D.; Schenke-Layland, K.; Oehr, C.; et al. Dose-Dependent Tissue-Level Characterization of a Medical Atmospheric Pressure Argon Plasma Jet. ACS Appl. Mater. Interfaces 2019, 11, 19841–19853. [Google Scholar] [CrossRef] [PubMed]
  52. Lee, J.-H.; Kim, K.-N. Effects of a Nonthermal Atmospheric Pressure Plasma Jet on Human Gingival Fibroblasts for Biomedical Application. Biomed Res. Int. 2016, 2016, 2876916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Jablonowski, L.; Kocher, T.; Schindler, A.; Müller, K.; Dombrowski, F.; von Woedtke, T.; Arnold, T.; Lehmann, A.; Rupf, S.; Evert, M.; et al. Side Effects by Oral Application of Atmospheric Pressure Plasma on the Mucosa in Mice. PLoS ONE 2019, 14, e0215099. [Google Scholar] [CrossRef] [Green Version]
  54. Evert, K.; Kocher, T.; Schindler, A.; Müller, M.; Müller, K.; Pink, C.; Holtfreter, B.; Schmidt, A.; Dombrowski, F.; Schubert, A.; et al. Repeated Exposure of the Oral Mucosa over 12 Months with Cold Plasma Is Not Carcinogenic in Mice. Sci. Rep. 2021, 11, 20672. [Google Scholar] [CrossRef]
  55. Jensen, J.-O.; Schulz, L.; Schleusser, S.; Matzkeit, N.; Stang, F.H.; Mailaender, P.; Kraemer, R.; Kleemann, M.; Deichmann, H.; Kisch, T. The Repetitive Application of Cold Atmospheric Plasma (CAP) Improves Microcirculation Parameters in Chronic Wounds. Microvasc. Res. 2021, 138, 104220. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Y.; Pan, J.; Ye, G.; Zhang, Q.; Wang, J.; Zhang, J.; Fang, J. In Vitro Studies of the Antimicrobial Effect of Non-Thermal Plasma-Activated Water as a Novel Mouthwash. Eur. J. Oral Sci. 2017, 125, 463–470. [Google Scholar] [CrossRef]
  57. Hoefeler, H.; Duran, I.; Hechmati, G.; Garzon Rodriguez, C.; Lüftner, D.; Ashcroft, J.; Bahl, A.; Atchison, C.; Wei, R.; Thomas, E.; et al. Health Resource Utilization Associated with Skeletal-Related Events in Patients with Bone Metastases: Results from a Multinational Retrospective—Prospective Observational Study—A Cohort from 4 European Countries. J. Bone Oncol. 2014, 3, 40–48. [Google Scholar] [CrossRef] [Green Version]
  58. Oliveira, A.; Simões, S.; Ascenso, A.; Reis, C.P. Therapeutic Advances in Wound Healing. J. Dermatol. Treat. 2022, 33, 2–22. [Google Scholar] [CrossRef] [PubMed]
  59. Kim, H.; Kim, H.-J.; Kim, H.K.; Hong, J.Y.; Cho, S.B. Effects of Argon and Nitrogen Plasma Pulses on the Skin and Skin Appendages in an in Vivo Animal Model. Skin Res. Technol. 2020, 26, 81–90. [Google Scholar] [CrossRef]
Figure 1. Influence of NIPP on Ki67 and PCNA mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) Ki67 mRNA in HGF; (b) Ki67 mRNA in HGK; (c) PCNA mRNA in HGF; (d) PCNA mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Figure 1. Influence of NIPP on Ki67 and PCNA mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) Ki67 mRNA in HGF; (b) Ki67 mRNA in HGK; (c) PCNA mRNA in HGF; (d) PCNA mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g001
Figure 2. Effects of NIPP on Ki67 protein levels in HGF and HGK at 24 h and 48 h, respectively. Protein levels were related to total protein and normalised to control. Treatment was performed for the indicated times: (a) Ki67 concentration in HGF cell lysates at 24 h; (b) Ki67 concentration in HGF cell lysates at 48 h; (c) Ki67 concentration in HGK cell lysates at 24 h; (d) Ki67 concentration in HGK cell lysates at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Figure 2. Effects of NIPP on Ki67 protein levels in HGF and HGK at 24 h and 48 h, respectively. Protein levels were related to total protein and normalised to control. Treatment was performed for the indicated times: (a) Ki67 concentration in HGF cell lysates at 24 h; (b) Ki67 concentration in HGF cell lysates at 48 h; (c) Ki67 concentration in HGK cell lysates at 24 h; (d) Ki67 concentration in HGK cell lysates at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g002
Figure 3. Impact of NIPP on COL1A1 and MMP1 mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) COL1A1 mRNA in HGF; (b) COL1A1 mRNA in HGK; (c) MMP1 mRNA in HGF; (d) MMP1 mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Figure 3. Impact of NIPP on COL1A1 and MMP1 mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) COL1A1 mRNA in HGF; (b) COL1A1 mRNA in HGK; (c) MMP1 mRNA in HGF; (d) MMP1 mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g003
Figure 4. Effects of NIPP on MMP1 protein levels in HGF and HGK at 24 h and 48 h, respectively. Protein levels were related to total protein and normalised to control. Treatment was performed for the indicated times: (a) MMP1 concentration in HGF cell culture supernatants at 24 h; (b) MMP1 concentration in HGF cell culture supernatants at 48 h; (c) MMP1 concentration in HGK cell culture supernatants at 24 h; (d) MMP1 concentration in HGK cell culture supernatants at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Figure 4. Effects of NIPP on MMP1 protein levels in HGF and HGK at 24 h and 48 h, respectively. Protein levels were related to total protein and normalised to control. Treatment was performed for the indicated times: (a) MMP1 concentration in HGF cell culture supernatants at 24 h; (b) MMP1 concentration in HGF cell culture supernatants at 48 h; (c) MMP1 concentration in HGK cell culture supernatants at 24 h; (d) MMP1 concentration in HGK cell culture supernatants at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g004
Figure 5. Influence of NIPP on CASP9 and CASP3 mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) CASP9 mRNA in HGF; (b) CASP9 mRNA in HGK; (c) CASP3 mRNA in HGF; (d) CASP3 mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Figure 5. Influence of NIPP on CASP9 and CASP3 mRNA levels in HGF and HGK at 24 h. Treatment was performed for the indicated times: (a) CASP9 mRNA in HGF; (b) CASP9 mRNA in HGK; (c) CASP3 mRNA in HGF; (d) CASP3 mRNA in HGK. n = 9. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g005
Figure 6. Effects of NIPP on Caspase-3/7 activity in HGF and HGK 24 h and 48 h after treatment, respectively. Data was normalised to control. Treatment was performed for the indicated times: (a) Caspase-3/7 activity in HGF at 24 h; (b) Caspase-3/7 activity in HGF at 48 h; (c) Caspase-3/7 activity in HGK at 24 h; (d) Caspase-3/7 activity in HGK at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Figure 6. Effects of NIPP on Caspase-3/7 activity in HGF and HGK 24 h and 48 h after treatment, respectively. Data was normalised to control. Treatment was performed for the indicated times: (a) Caspase-3/7 activity in HGF at 24 h; (b) Caspase-3/7 activity in HGF at 48 h; (c) Caspase-3/7 activity in HGK at 24 h; (d) Caspase-3/7 activity in HGK at 48 h. n = 6. * statistical significance compared to control (0 s) (p < 0.05).
Biomedicines 10 00889 g006
Figure 7. Effects of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). Hematoxylin and eosin (HE) staining, original magnification 10×. The scale bar represents 250 µm.
Figure 7. Effects of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). Hematoxylin and eosin (HE) staining, original magnification 10×. The scale bar represents 250 µm.
Biomedicines 10 00889 g007
Figure 8. Influence of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). Ki67 staining, original magnification 10× and 40×. Blue arrows: positively stained HGK, black arrows: positively stained HGF. The scale bar represents 250 µm.
Figure 8. Influence of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). Ki67 staining, original magnification 10× and 40×. Blue arrows: positively stained HGK, black arrows: positively stained HGF. The scale bar represents 250 µm.
Biomedicines 10 00889 g008
Figure 9. Influence of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). MMP1 staining, original magnification 10× and 40×. Red arrows: positively stained HGK, black arrows: positively stained HGF. The scale bar represents 250 µm.
Figure 9. Influence of NIPP on gingival tissue biopsies at 24 h after NIPP treatment for 30 s and 60 s, as compared to the untreated group (NIPP 0 s). MMP1 staining, original magnification 10× and 40×. Red arrows: positively stained HGK, black arrows: positively stained HGF. The scale bar represents 250 µm.
Biomedicines 10 00889 g009
Figure 10. Markers of proliferation and remodeling were upregulated by NIPP in HGF, HGK, and tissue biopsies.
Figure 10. Markers of proliferation and remodeling were upregulated by NIPP in HGF, HGK, and tissue biopsies.
Biomedicines 10 00889 g010
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Eggers, B.; Stope, M.B.; Marciniak, J.; Götz, W.; Mustea, A.; Deschner, J.; Nokhbehsaim, M.; Kramer, F.-J. Non-Invasive Physical Plasma Generated by a Medical Argon Plasma Device Induces the Expression of Regenerative Factors in Human Gingival Keratinocytes, Fibroblasts, and Tissue Biopsies. Biomedicines 2022, 10, 889. https://doi.org/10.3390/biomedicines10040889

AMA Style

Eggers B, Stope MB, Marciniak J, Götz W, Mustea A, Deschner J, Nokhbehsaim M, Kramer F-J. Non-Invasive Physical Plasma Generated by a Medical Argon Plasma Device Induces the Expression of Regenerative Factors in Human Gingival Keratinocytes, Fibroblasts, and Tissue Biopsies. Biomedicines. 2022; 10(4):889. https://doi.org/10.3390/biomedicines10040889

Chicago/Turabian Style

Eggers, Benedikt, Matthias Bernhard Stope, Jana Marciniak, Werner Götz, Alexander Mustea, James Deschner, Marjan Nokhbehsaim, and Franz-Josef Kramer. 2022. "Non-Invasive Physical Plasma Generated by a Medical Argon Plasma Device Induces the Expression of Regenerative Factors in Human Gingival Keratinocytes, Fibroblasts, and Tissue Biopsies" Biomedicines 10, no. 4: 889. https://doi.org/10.3390/biomedicines10040889

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop