Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

Devitalization of Glioblastoma Cancer Cells by Non-invasive Physical Plasma: Modulation of Proliferative Signalling Cascades

SEBASTIAN LEHMANN, SANDRA BIEN-MÖLLER, SASCHA MARX, SANDER BEKESCHUS, HENRY W.S. SCHROEDER, ALEXANDER MUSTEA and MATTHIAS B. STOPE
Anticancer Research January 2023, 43 (1) 7-18; DOI: https://doi.org/10.21873/anticanres.16128
SEBASTIAN LEHMANN
1Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SANDRA BIEN-MÖLLER
1Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany;
2Department of Pharmacology, University Medicine Greifswald, Greifswald, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SASCHA MARX
3Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SANDER BEKESCHUS
4ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HENRY W.S. SCHROEDER
1Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ALEXANDER MUSTEA
5Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MATTHIAS B. STOPE
5Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: matthias.stope@ukbonn.de
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Glioblastoma (GBM) is the most common and most lethal type of cancer of the central nervous system in adults. Despite aggressive treatment, which is based on surgical resection, if possible, followed by radiation and chemotherapy, a high recurrence rate and therapy resistance is observed. Thus, additional innovative therapies are urgently needed to improve the poor median survival of only 15 months. Treatment of solid tumours with non-invasive physical plasma (NIPP) represents such a novel and innovative anticancer procedure. Materials and Methods: In this study, we investigated the effect of NIPP, an ionized argon gas, on the in vitro growth of human GBM cell lines, LN-18 and U-87 MG. Proliferation was measured by live cell count. Subsequently, proliferative factors were analysed at the level of nucleic acids (polymerase chain reaction) and proteins (western blotting). Results: For both GBM lines, a treatment time-dependent decrease in growth was observed compared to controls. Additionally, NIPP treatment resulted in reduced rates of AKT serine/threonine kinase 1 (AKT1) and extracellular-regulated kinase 1/2 ERK1/2 expression, whereas expression of p21, proliferating cell nuclear antigen, and heat-shock proteins 90α and 90β was not affected. In both cell lines, a strong increase in expression of tumour-suppressive microRNA-1 (miR-1) was detected after exposure to NIPP. Conclusion: Our results demonstrated that NIPP is able to efficiently attenuate growth of GBM cells and suggest AKT1, ERK1/2 and miR-1 to be pivotal factors of NIPP-modulated cellular signalling. Translated into the clinical setting, NIPP may represent a promising option for the treatment of GBM.

Key Words:
  • Cold atmospheric plasma
  • cold atmosphere pressure plasma
  • tissue tolerable plasma
  • microRNA-1
  • AKT1

Glioblastoma (GBM) represents the most aggressive brain tumour of glial origin and accounts for approximately 60% of all brain-derived tumours. The invasive and nearly always lethal phenotype of GBM is particularly characterized by behaviour of rapid and infiltrative growth deep into the healthy brain tissue (1-3). Despite multimodal therapy consisting of surgical resection followed by chemotherapy and radiation, the median survival is still only about 15 months. In spite of the investigation of new GBM treatment approaches, no substantial improvement in GBM therapy has been achieved in recent years (4, 5).

A promising and novel therapy option is the treatment of tumours with non-invasive physical plasma (NIPP), previously also known as cold atmospheric (pressure) plasma, which primarily consists of an ionized gas. Unlike classical thermal plasma, the fourth state of matter, NIPP is not in a thermal equilibrium, the ions in the gas cloud have a much lower kinetic energy than the ions in classical thermal plasma. This makes it possible to generate the plasma at room temperature (6). NIPP is generated by high-frequency, high-voltage discharges between two electrodes. The high energy input ionizes the gas passing the electrodes and NIPP is formed (6). Currently, there are two main techniques used to generate NIPP. In the plasma jet principle, a gas, usually a noble gas, is ionized between two electrodes and emerges as NIPP effluent. In dielectric barrier discharge, on the other hand, the ambient atmosphere between the electrode and the grounded surface to be treated is used to form a NIPP (6-8).

According to the current understanding, the effect of NIPP is based primarily on the formation of reactive oxygen species (ROS) which interact with cellular structures (9). ROS are formed at the interface of plasma and ambient atmosphere. In tissue, ROS lead to the induction of oxidative stress in the extracellular matrix, as well as in intracellular compartments. Due to the application of NIPP, modifications of the extracellular matrix, the cell membrane, and intracellular components and structures are observed. In particular, changes in cytoplasmic membrane permeability and induction of DNA double-strand breaks have been detected (9, 10). Thereby, dose-dependent effects of NIPP have been described for some time. At low doses, proliferative and pro-angiogenic actions of NIPP have been demonstrated (10). At high doses, cell damage resulting in apoptosis and necrosis was found. In this regard, not only were antimicrobial effects detected, but also a strong selectivity of NIPP in impairing the growth of tumour cells was shown (6, 8, 9). A large number of potential applications of NIPP are currently in clinical and pre-clinical investigation. At present, most advanced is the research regarding the application of NIPP for the treatment of acute and chronic wounds. Other possible areas of application which are currently under research include dentistry, ophthalmology, and the large field of oncology (8, 9, 11).

Although some studies have investigated the effects of NIPP on GBM, the underlying molecular mechanisms are still poorly understood. We therefore evaluated the effect of NIPP treatment on an in vitro GBM model, consisting of the well-established cell lines LN-18 and U-87 MG. Molecular analyses included of growth and resistance-associated proteins serine/threonine kinase 1 (AKT1), p44/p42 mitogen-activated protein kinase (ERK1/2), cyclin-dependent kinase inhibitor 1 (p21), proliferating cell nuclear antigen (PCNA), heat-shock protein 90 (HSP90) and microRNA-1 (miR-1).

Materials and Methods

Cell culture and NIPP treatment. The maintenance of LN-18 and U-87 MG cells (both human GBM cell lines authenticated by the American Type Culture Collection, Manassas, VA, USA) was performed in Dulbecco’s modified Eagle’s medium supplemented with 10% foetal bovine serum, 2 mM glutamine and 1% non essential amino acid solution (100×) (all from PAN Laboratories, Cölbe, Germany) at 37°C with 95% humidity and 5% CO2.

For NIPP treatment, we used a Kinpen 09, a plasma jet manufactured by Neoplas tools (Greifswald, Germany). This physical plasma jet is operated with argon as carrier gas at a flow rate of 3 l/min. It creates a 9-13 mm long plasma effluent with a temperature below 42°C, and the sinusoidal operating frequency is approximately 1 MHz (12).

For cell growth experiments, 15,000 cells/well were seeded into a 24-well cell culture plate. In the experimental group, treatment with NIPP was performed for 5 s or 15 s. The control group was treated with non-ionized argon for the same time period. The treatment of cells with the Kinpen 09 jet was performed manually. The jet was placed at a distance of 9-13 mm above the cell suspension such that the tip of the effluent only reached the surface of the cell culture medium. Subsequently, the cells were transferred into new 24-well plates in order to exclude a possible influence of NIPP on the surface of the primarily used cell culture plate. Cell growth was determined after 4, 24, 48, 72, 96, and 120 h of incubation using a CASY TT cell counter and analyser (Roche Applied Science, Mannheim, Germany) with a 150 μm capillary, after detaching the cells with trypsin. The measurement was performed three times with 400 μl each in triplicate.

Western blotting. For analysis of the expression and phospho-activation of selected proliferation-related proteins (AKT1, ERK1/2, HSP90α, HSP90β, p21, and PCNA), the treatment time with NIPP was set to 15 s. Measurement of target proteins was performed after 24 and 48 h of incubation. For preparation of protein lysates, harvested cells were incubated on ice for 30 min with lysis buffer (50 mM Tris-HCl pH 7.4, 100 mM NaCl, 0.1% Triton X-100, 5 mM EDTA, 1 mM leupeptin, 1 mM aprotinin and 250 μg/ml sodium vanadate). BCA Protein Assay Kit (Thermo Fisher Scientific, Rockford, IL, USA) was used to determine protein concentration of cell lysates. After protein denaturation in sodium dodecyl sulphate (SDS) sample buffer (50 mM Tris-HCl pH 6.8, 2% SDS, 10% glycerol, 0.01% bromophenol blue, 5% 2-mercaptoethanol) at 95°C for 5 min, 30 μg of each protein sample were separated on an SDS polyacrylamide gel. For immunoblotting a tank blot system (Bio-Rad, Hempstead, UK) was used to transfer separated proteins onto a nitrocellulose membrane (GE Healthcare Life Science, Solingen, Germany). After blotting, membranes were blocked in 5% fetal bovine serum in Tris-buffered saline containing 0.05% Tween 20 (TBST) and 1% bovine serum albumin for 1 h at room temperature. For immunodetection, membranes were incubated with the primary antibodies (Table I) overnight at 4°C followed by a washing procedure with TBST. Incubation with the secondary antibodies was performed for one hour at room temperature again followed by washing step with TBST. For detection of chemiluminescent signals of horseradish peroxidase-coupled secondary antibodies, ChemiDoc XRS Imaging System (Bio-Rad) using ECL Plus Western Blotting Substrate (Thermo Fisher Scientific, Waltham, MA, USA) followed by densitometric analysis (Quantity One; Bio-Rad) was used. Fluorescence signals of IRDye-conjugated secondary antibodies were detected using an Odyssey CLx Imaging System (Li-cor Bioscience, Bad Homburg, Germany). The relative chemiluminescence or fluorescence intensities of the specific bands were calculated and normalized to those of glyceraldehyde 3-phosphate dehydrogenase as a loading control.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Antibodies for western blot analysis.

Quantitative reverse transcription-polymerase chain reaction (RT-PCR). RT-PCR was used for quantification of miR-1 expression after treatment of GBM cells with NIPP for 15 s, followed by a cell culture period of 24 and 48 h. For RNA isolation by phase segregation, peqGOLD TriFast (Peqlab, Erlangen, Germany) was used according to the manufacturer’s protocol. Total RNA was determined by NanoDrop 2000 spectrometer (Peqlab), and samples were adjusted to a content of 100 ng RNA/μl. Synthesis of cDNA coding for miR-1 and the housekeeping gene U6 was achieved by a Thermal Cycler T3000 (Biometra, Göttingen, Germany) performing stem-loop RT-PCR with primers targeting specifically miR-1 and U6 (miR-1 SLOOP, U6 SLOOP, Table II).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Oligonucleotides for quantitative reverse transcription-polymerase chain reaction analysis.

Nucleic acid quantification was performed by real-time PCR with a CFX96 Real-Time PCR Detection System (Bio-Rad). Amplification of miR-1 and U6-coding cDNA was achieved using specific forward- and reverse primers. Simultaneous fluorescence-based quantification was performed via SYBR-Green detection using SensiMix SYBR Hi-ROX Kit (Bioline, Luckenwalde, Germany) between the amplification cycles. The analysis of the quantitative PCR data was performed in CFX Manager 2.0 (Bio-Rad).

Statistics. Statistical analysis and graphical representation were performed using GraphPad Prism software 5.0 (GraphPad Software, Inc., San Diego CA, USA). Analysis of variance served as the test procedure for cell kinetics, and the Wilcoxon signed-rank test was used for analyses of western blot and RT-PCR data. A value of p<0.05 was considered to be statistically significant. Grubbs’ test for identification of outliers was performed using GraphPad QQuickCalcs Outlier calculator.

Results

Dose- and time-dependent growth-inhibitory effect of NIPP on GBM cells. In our study, we first evaluated the effect of NIPP on GBM cell proliferation by performing growth kinetics based on the measurement of living cells at 4, 24, 48, 72, 96 and 120 h after NIPP treatment. Both NIPP treatment durations led to a significant reduction in growth rate compared to the control group in U-87 MG cells (Figure 1A), as well as in LN-18 cells (Figure 1B). This growth-inhibitory effect was most pronounced after 72, 96 and 120 h. In addition, there was a clear increase in growth inhibition when the duration of NIPP treatment was increased from 5 to 15 s. This observation was consistent and similar in both GBM cell lines. The LN-18 growth kinetics showed 48% less cell growth after 5 s of treatment, while after 15 s of treatment, a decrease of 87% was observed. The results for U-87 MG matched these findings, with growth rate reductions of 44% and 69%, respectively.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Comparison of the effects of non-invasive physical plasma (NIPP) on cell growth of U-87 MG (A, B) and LN-18 (C, D) cells. Cells were treated for 5 (A, C) or 15 s (B, D) with NIPP or the carrier gas (argon; control). Living cells were assessed applying a CASY Cell Counter and Analyzer (OLS, Bremen, Germany) at 4, 24, 48, 72, 96, and 120 h after treatment. Data are given as mean and standard deviation (U-87 MG n=6, LN-18 n=5), two-way ANOVA with Bonferroni post-hoc test. Significantly different at: p<0.05; **p≤0.01; ***p≤0.001.

Reduction of the AKT1 and ERK1/2 protein expression after NIPP treatment. To evaluate which signalling pathways might be affected by NIPP treatment, we analysed the expression and phospho-activation of two proliferation-related kinases: AKT1 and ERK1/2 (13, 14). Figure 2 represents the expression and activation data of both AKT1 and ERK1/2 in U-87 MG (Figure 2A) and LN-18 (Figure 2B) cells.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Western blot analyses of total (t-) and phosphorylated (p-) forms of serine/threonine kinase 1 (AKT1) and extracellular-regulated kinase 1/2 (ERK1/2) in U-87 MG (A) and LN-18 (B) glioblastoma cells. Cells were treated for 15 s with non-invasive physical plasma (NIPP) or carrier gas (argon; control) and protein expression was determined 24 and 48 h after NIPP treatment. Expression of t-AKT1 and t-ERK1/2 was normalized to the level of the housekeeping protein glyceraldehyde 3-phosphate dehydrogenase (GAPDH). p-AKT1 and p-ERK1/2 were normalized to the content of t-AKT1 and t-ERK1/2, respectively. The upper panels show representative blots in each case; the lower panels present the statistical analyses of 4-8 independent experiments as the mean (relative to that of control cells) and standard deviation. Significantly different at: *p<0.05 and **p≤0.01 by Wilcoxon signed-rank test.

U-87 MG cells showed a significant decrease (by half) in the expression of total AKT1 24 h after treatment with NIPP (Figure 2A). At the same time, the amount of phosphorylated, active AKT1 (p-AKT) was significantly increased (1.83-fold) by NIPP treatment (Figure 2B). However, 48 h after NIPP treatment, the relative levels of both activated and total AKT1 had returned to normal in U-87 MG cells. A similar pattern was observed for total ERK1/2 and its phosphorylated form (p-ERK1/2), with reduction of total ERK expression to 0.68 and a 2.77-fold increase in active p-ERK1/2 24 h after NIPP treatment of U-87 MG cells (Figure 2A), but this was not statistically significant (p=0.0625).

In LN-18 cells, on the other hand, there was a slight (1.5- to 2.0-fold) but not significant increase in both the total and active AKT1 level after NIPP treatment. The total EKR1/2 expression was not significantly changed by NIPP but p-ERK1/2 was slightly reduced to 0.78 48 h after NIPP treatment of LN-18 cells (Figure 2B).

Expression of HSP90α, HSP90β, p21, and PCNA is not affected after NIPP treatment. Since it is well-known that HSP90 promotes GBM cell viability by interaction with a wide range of client proteins to influence growth, survival, and the cell cycle (15), we next analysed HSP90α and HSP90β expression at 24 and 48 h after NIPP treatment. As shown in Figure 3, in both U-87 MG (Figure 3A) and LN-18 (Figure 3B) cells, no significant change in either HSP90α or HSP90β expression was detectable. Furthermore, protein expression of p21 and PCNA was also not significantly changed, despite a trend for a reduced PCNA content in U-87 MG cells 48 h after NIPP treatment.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Western blot analyses of heat-shock protein 90α (HSP90α), HSP90β, p21, and proliferating cell nuclear antigen (PCNA) in U-87 MG (A) and LN-18 (B) glioblastoma cells. Cells were treated for 15 s with non-invasive physical plasma (NIPP) or carrier gas (argon; control) and protein expression was determined 24 and 48 h after NIPP treatment. Expression of proteins was normalized to the level of the housekeeping protein glyceraldehyde 3-phosphate dehydrogenase (GAPDH). The upper panels show representative blots in each case; the lower panels present the statistical analyses of 4-8 independent experiments as the mean (relative to that of control cells) and standard deviation. There were no significant differences by Wilcoxon signed-rank test.

Expression of tumour-suppressive miR-1 is elevated after NIPP treatment. An additional regulator of tumour progression is the tumour suppressor miR-1. As shown in Figure 4, 24 h after NIPP treatment a significant, almost 15-fold increase of the miR-1 level in U-87 MG cells and a more than 5-fold up-regulation in LN-18 cells was observed. Interestingly, 48 h after NIPP treatment, a decrease of the miR-1 content to the level of the controls was detectable in both GBM cell lines (Figure 4).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Expression of miR-1 in U-87 MG (left) and LN-18 (right) glioblastoma cells. Cells were treated for 15 s with non-invasive physical plasma (NIPP) or carrier gas (argon; control) and miR-1 expression was determined 24 and 48 h after NIPP treatment using quantitative reverse transcription-polymerase chain reaction analysis. miR-1 expression was normalized to that of endogenously expressed U6 snRNA. Data are given as the mean and standard deviation (n=5-7). *Significantly different at p<0.05 by Wilcoxon signed-rank test.

Discussion

GBM is one of the most deadly and difficult-to-treat brain cancers because of its infiltrative growth behaviour. Complete tumour removal by surgery is nearly impossible. Due to therapy resistance, patients with GBM experience relapse within a few months, resulting in a short median overall survival of only about 15 months. This emphasizes the urgent need for novel GBM therapies. NIPP has emerged as potential and innovative anti-tumoural option for solid tumours.

NIPP has been investigated for its suitability in several applications, including the treatment of cancer (16, 17). The treatment of cancer has diversified with the use of NIPP, which was shown to induce apoptosis in many types of cancers, including melanoma, pancreatic, ovarian, breast cancer or lung cancer (18-22). Some studies evaluating the tumour-suppressive effect of NIPP on GBM cells also exist (23-26), however, the influence of NIPP on the regulation of intracellular signalling cascades and growth-regulatory proteins is poorly understood. In in vitro experiments, an increased ROS content, intracellularly and extracellularly, as well as a restoration of sensitivity of O6-methyl-guanine DNA methyltransferase-positive tumour cells to temozolomide was demonstrated (6, 27, 28). More importantly, the first in vivo study also showed a significant reduction in growth of GBM in mice when treated with NIPP (26).

In this study, we investigated the influence of NIPP treatment on growth of two human GBM cell lines, LN-18 and U-87 MG. Additionally, we analysed the expression of key intracellular factors (AKT1, ERK1/2, HSP90α, HSP90β, p21, PCNA and miR-1) which are known to be relevant for GBM growth and progression.

In our study, we demonstrated a significant growth-inhibitory effect of NIPP on GBM cell models. Such an anti-tumoural impact of NIPP was already described in the literature using different GBM cell lines and affirms the potential application of NIPP as a therapeutic option for treatment of patients with GBM (23-25, 28, 29). As partly shown by others, we also confirm the dose- and time-dependent effects of NIPP treatment. The earliest significant reductions in cell number were found 72 h after NIPP application to LN-18 and U-87 MG cells, an effect which was much stronger after 96 and 120 h of incubation. Additionally, treatment of GBM cells for 15 s was more effective than that for 5 s. The strong growth-inhibitory effects of NIPP treatment were associated with a regulation of signalling molecules relevant for tumour growth and progression which will be discussed in detail below.

NIPP-induced growth suppression was accompanied by a significant change in the regulation of the pro-oncogenic survival kinase AKT1, which is an effector kinase of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)-AKT pathway. Strong evidence has revealed that AKT1 also plays a major role in GBM progression and aggressiveness (30). AKT1 is a key protein associated with anti-apoptotic processes, proliferation, and migration (31). In many tumour types, including GBM, AKT1 expression has been found to be elevated during progression. Thus, AKT1 is discussed as a crucial factor in tumourigenesis (13). We found a decrease in total AKT1 expression in U-87 MG cells 24 h after NIPP treatment. Due to its important role in cell survival, this reduction of AKT1 expression may be directly caused by NIPP-mediated cytotoxic effects. But unexpectedly, the amount of phosphorylated, active AKT1 was increased 24 h after NIPP treatment. Whether this was actually an increase in the activity of AKT1 or only a relative excess due to the reduced total AKT1 content cannot be answered by our experiments. Furthermore, 48 h after NIPP treatment, the levels of both total and p-AKT1 returned to those of the control. This may mean that NIPP causes a short-term change in regulation of AKT1 which might be sufficient to induce other growth-regulatory cascades. The temporary increase in active AKT1 protein might also represent a rapidly activated survival cascade to protect GBM cells from death, which then cannot be maintained due to further cell damage processes caused by NIPP. Interestingly, in LN-18 cells, no significant change in expression of AKT1 by NIPP was found in our study, but there was a trend for increased total and active AKT1. Discrepancies between LN-18 and U-87 MG cells might be based on the different status of phosphatase and tensin homologue (PTEN), which is negatively regulates the activation of AKT1 (32). While LN-18 express wild-type PTEN, U-87 MG cells are described as PTEN-negative, which might be the reason for the observed significant increase in active AKT1 in U-87 MG caused by the lack of PTEN-mediated inactivation of AKT1 (33). At later time points, it is conceivable that other factors control AKT1 activation such as integrin-linked kinase, which has been shown to regulate AKT1 activity in PTEN-mutant GBM cells (34, 35). Regulation of AKT1 by NIPP is not described in the literature, but oxidative stress, which is thought to be induced by NIPP, was shown to inactivate PI3K/AKT1 signalling, while activity of its negative regulator PTEN appears not to be regulated directly (36, 37).

Besides AKT1, a role of ERK1/2 in GBM pathogenesis is discussed (38). A trend for a similar pattern of regulation was found for ERK1/2, which also belongs to the survival kinases, in U-87 MG cells. Again, there was a slight down-regulation of total ERK1/2 protein but an enhanced level of ERK1/2 phosphorylation 24 h after NIPP treatment. An increase in ERK1/2 phospho-activation has been already shown after treatment of melanoma cells and myeloblasts with NIPP (39, 40). In contrast, reduced total and active ERK1/2 were found in NIPP-treated prostate cancer cells (41). Thus, modulation of ERK1/2 by NIPP seems to be cell type-dependent. As a component of the RAS-ERK signalling cascade, ERK1/2 is an oncogenic kinase capable of influencing a variety of effectors, which are particularly associated with anti-apoptotic processes, cell growth and tumour progression (42, 43). Thus, an increased short-term activation of ERK1/2 by NIPP may represent a mode of protection for GBM cells to escape cell death as described for AKT1 above, which, however, cannot be maintained by additional cellular stress processes. Again, the observed changes of ERK1/2 in U-87 MG cells were not found in LN-18 cells. In contrast, there was a slight, but significant reduction in the level of p-ERK1/2 in LN-18 cells at 48 h after NIPP treatment. Once more, the different PTEN status of U-87 MG (PTEN-mutant) and LN-18 (PTEN wild-type) might be responsible for the different response of these two GBM cells to NIPP. Intact PTEN, described for LN-18 cells, is able to reduce ERK1/2 activation through inhibition of RAS, which can counteract the activation of ERK1/2 in LN-18 cells (44). In PTEN-mutant U-87 MG cells, there is no inhibition of RAS and thus also not of ERK1/2, which may conceivably lead to a greater increase in ERK1/2 activity.

Both the PI3K/AKT1 and mitogen-activated protein kinase/ERK1/2 pathways, as prominent drivers of GBM cell proliferation, are strongly dependent on HSP90 protein (15). Under chemotherapy, HSP90 promotes GBM cell viability and stabilizes inhibitors of apoptotic mechanisms. Thus, HSP90 contributes to chemoresistance (45, 46). Generally, HSP90 is considered a molecular chaperone responsible for correct folding, stabilization, and transport of proteins (47, 48). Two HSP90 isoforms are mainly expressed: the stress-induced HSP90α and the constitutively expressed HSP90β type (47). In many tumours, including GBM, expression of HSP90 was observed to be elevated (45, 49). Numerous studies have demonstrated that inhibition of HSP90 reduces the growth of GBM cells (50, 51). However, tumour-selective cleavage of HSP90 by oxidative stress and NIPP have also been reported (52, 53), but no data exist on GBM regarding regulation of HSP90 by NIPP or oxidative stress as far as we are aware. In our study, no significant changes of HSP90 isoforms α and β were detected in LN-18 and U-87 MG cells after NIPP application. Thus, HSP90 appears not to be a target of NIPP-mediated growth-inhibitory effects in GBM cells.

Both apoptosis and cell-cycle arrest are found to be induced by NIPP in tumour cells (54, 55). Expression analyses of the apoptosis-related and cell cycle-modulating proteins p21 and PCNA, which are also significantly involved in DNA replication and DNA repair, also showed no significant changes after NIPP application in both LN-18 and U-87 MG cells (56, 57). Due to the growth-inhibitory effects of NIPP, a change in expression of the cell-cycle regulator p21 after NIPP treatment would have been conceivable. In contrast to our results, melanoma, colorectal and prostate cancer cells treated with NIPP demonstrated a significant modulation of p21 (41, 58). This argues for a cell type-dependent regulation of p21 expression after NIPP exposure and may link to p21-independent growth inhibition in GBM cells. Regarding PCNA, a cell type-specific regulation was found after NIPP treatment. Whereas an increase of PCNA expression was described in human gingival and osteoblast-like cells (59, 60), modulation of PCNA was lacking in squamous cell carcinoma and malignant melanoma cells (61). In line with these findings, analysis of PCNA expression in our study also revealed a lack of change in PCNA expression after NIPP treatment of GBM cells. PCNA is responsible for a fast and smooth process of DNA synthesis and the initiation of DNA repair processes (59). There is currently much discussion whether NIPP-induced ROS directly cause DNA damage or only induce apoptosis or other forms of cell death (62).

miRs play pivotal roles in tumourigenesis, apoptosis, and drug resistance in cancer cells due to their regulatory function in post-transcriptional gene expression (62). Among them, the tumour-suppressive miR-1 has been shown to be predominantly down-regulated in almost all human cancer types, including GBM (62, 63). miR-1 suppresses the expression of factors that promote cell growth and angiogenesis and suppress apoptosis (64). To the best of our knowledge, this is the first study to demonstrate a strong increase of miR-1 after NIPP application in both LN-18 and U-87 MG cells. Thus, miR-1 up-regulation may represent a molecular mediator responsible for the tumour-suppressive effects of NIPP. Remarkably, the increase in miR-1 expression was only temporary and expression returned to the control level after 48 h of incubation, however, this short-term induction in miR-1 expression might be sufficient for the subsequent signal cascades to inhibit cell growth. Moreover, miR-1 has been classified as having a high potential to inhibit tumour progression and therapy resistance (65). Recently, Bronisz and colleagues showed that ectopic expression of miR-1 in GBM cells inhibited cell growth, neovascularisation, and invasiveness in vivo (63). miR-1 seems to have an important role as an effector molecule for inhibition of the aggressive growth behaviour of tumour cells such as GBM.

Conclusion

NIPP represents a new and promising treatment option for therapy of a variety of tumours, including GBM. Although most studies are still in the experimental stage, there are initial studies that demonstrate benefits of its application in patients. Nevertheless, there still seems to be a long way to go before clinical NIPP application can find its way into routine oncological therapy. Of note, a challenging part of treating GBM is that it is present in the brain, and any damage to the tissue during surgery or other treatment can cause a series of devastating effects in the central nervous system. Thus, the non-invasive set-up of NIPP, as suggested by Soni and colleagues (67), would be an important improvement for therapeutic options.

To date, many of the intracellular processes induced by NIPP in tumour cells have not been adequately studied or understood. Our study argues for a possible role of kinases, and especially of miR-1, as potential effectors of the growth-inhibitory effect of NIPP. This should be the subject of further research to improve the extremely poor survival of patients with GBM.

Acknowledgements

The Authors thank Lena Zeyda for her dedicated experimental support and Christian Scharf for his interested guidance of the NIPP experiments.

Footnotes

  • Authors’ Contributions

    Conceptualization, S.B.M., M.B.S., S.L.; methodology, S.L., S.M., S.B.; formal analysis, S.L., S.B.M., S.B. and M.B.S.; investigation, S.L., S.B.M., S.M., S.B. and M.B.S.; writing - original draft preparation, S.L., S.B.M., H.W.S.S., A.M. and M.B.S.; writing - review and editing, S.L., S.B.M., H.W.S.S., A.M. and M.B.S.; visualization, S.L., S.B.M. and M.B.S.; supervision S.B.M. and M.B.S.

  • Conflicts of Interest

    The Authors declare that they have no competing interests.

  • Received October 8, 2022.
  • Revision received November 10, 2022.
  • Accepted November 14, 2022.
  • Copyright © 2023 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Rock K,
    2. McArdle O,
    3. Forde P,
    4. Dunne M,
    5. Fitzpatrick D,
    6. O’Neill B and
    7. Faul C
    : A clinical review of treatment outcomes in glioblastoma multiforme – the validation in a non-trial population of the results of a randomised Phase III clinical trial: has a more radical approach improved survival? Br J Radiol 85(1017): e729-e733, 2012. PMID: 22215883. DOI: 10.1259/bjr/83796755
    OpenUrlAbstract/FREE Full Text
    1. Lukas RV,
    2. Wainwright DA,
    3. Ladomersky E,
    4. Sachdev S,
    5. Sonabend AM and
    6. Stupp R
    : Newly diagnosed glioblastoma: a review on clinical management. Oncology (Williston Park) 33(3): 91-100, 2019. PMID: 30866031.
    OpenUrlPubMed
  2. ↵
    1. Agnihotri S,
    2. Burrell KE,
    3. Wolf A,
    4. Jalali S,
    5. Hawkins C,
    6. Rutka JT and
    7. Zadeh G
    : Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies. Arch Immunol Ther Exp (Warsz) 61(1): 25-41, 2013. PMID: 23224339. DOI: 10.1007/s00005-012-0203-0
    OpenUrlCrossRefPubMed
  3. ↵
    1. Taylor OG,
    2. Brzozowski JS and
    3. Skelding KA
    : Glioblastoma multiforme: an overview of emerging therapeutic targets. Front Oncol 9: 963, 2019. PMID: 31616641. DOI: 10.3389/fonc.2019.00963
    OpenUrlCrossRefPubMed
  4. ↵
    1. Bianco J,
    2. Bastiancich C,
    3. Jankovski A,
    4. des Rieux A,
    5. Préat V and
    6. Danhier F
    : On glioblastoma and the search for a cure: where do we stand? Cell Mol Life Sci 74(13): 2451-2466, 2017. PMID: 28210785. DOI: 10.1007/s00018-017-2483-3
    OpenUrlCrossRefPubMed
  5. ↵
    1. Yan D,
    2. Sherman JH and
    3. Keidar M
    : Cold atmospheric plasma, a novel promising anti-cancer treatment modality. Oncotarget 8(9): 15977-15995, 2017. PMID: 27845910. DOI: 10.18632/oncotarget.13304
    OpenUrlCrossRefPubMed
    1. Weltmann KD,
    2. Kindel E,
    3. Woedtke T,
    4. Hähnel M,
    5. Stieber M and
    6. Brandenburg R
    : Atmospheric pressure plasma sources: prospective tools for plasma medicine. Pure Appl Chem 82: 1223-1237, 2010. DOI: 10.1351/PAC-CON-09-10-35
    OpenUrlCrossRef
  6. ↵
    1. Woedtke T,
    2. Emmert S,
    3. Metelmann HR,
    4. Rupf S and
    5. Weltmann KD
    : Perspectives on cold atmospheric plasma (CAP) applications in medicine. Phys Plasmas 27: 70601, 2020. DOI: 10.1063/5.0008093
    OpenUrlCrossRef
  7. ↵
    1. Semmler ML,
    2. Bekeschus S,
    3. Schäfer M,
    4. Bernhardt T,
    5. Fischer T,
    6. Witzke K,
    7. Seebauer C,
    8. Rebl H,
    9. Grambow E,
    10. Vollmar B,
    11. Nebe JB,
    12. Metelmann HR,
    13. Woedtke TV,
    14. Emmert S and
    15. Boeckmann L
    : Molecular mechanisms of the efficacy of cold atmospheric pressure plasma (CAP) in cancer treatment. Cancers (Basel) 12(2): 269, 2020. PMID: 31979114. DOI: 10.3390/cancers12020269
    OpenUrlCrossRefPubMed
  8. ↵
    1. Haertel B,
    2. von Woedtke T,
    3. Weltmann KD and
    4. Lindequist U
    : Non-thermal atmospheric-pressure plasma possible application in wound healing. Biomol Ther (Seoul) 22(6): 477-490, 2014. PMID: 25489414. DOI: 10.4062/biomolther.2014.105
    OpenUrlCrossRefPubMed
  9. ↵
    1. Stope MB
    : Plasma oncology – physical plasma as innovative tumor therapy. J Cancer Biol 1: 53-56, 2020. DOI: 10.46439/cancerbiology.1.010
    OpenUrlCrossRef
  10. ↵
    1. Reuter S,
    2. Woedtke T and
    3. Weltmann KD
    : The Kinpen–a review on physics and chemistry of the atmospheric pressure plasma jet and its applications. J Phys D Appl Phys 51: 233001, 2018. DOI: 10.1088/1361-6463/aab3ad
    OpenUrlCrossRef
  11. ↵
    1. McDowell KA,
    2. Riggins GJ and
    3. Gallia GL
    : Targeting the AKT pathway in glioblastoma. Curr Pharm Des 17(23): 2411-2420, 2011. PMID: 21827416. DOI: 10.2174/138161211797249224
    OpenUrlCrossRefPubMed
  12. ↵
    1. Roux PP and
    2. Blenis J
    : ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68(2): 320-344, 2004. PMID: 15187187. DOI: 10.1128/MMBR.68.2.320-344.2004
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. van Ommeren R,
    2. Staudt MD,
    3. Xu H and
    4. Hebb MO
    : Advances in HSP27 and HSP90-targeting strategies for glioblastoma. J Neurooncol 127(2): 209-219, 2016. PMID: 26842818. DOI: 10.1007/s11060-016-2070-8
    OpenUrlCrossRefPubMed
  14. ↵
    1. Laroussi M
    : Cold plasma in medicine and healthcare: The new frontier in low temperature plasma applications. Frontiers in Physics 8, 2020. DOI: 10.3389/fphy.2020.00074
    OpenUrlCrossRef
  15. ↵
    1. Keidar M,
    2. Walk R,
    3. Shashurin A,
    4. Srinivasan P,
    5. Sandler A,
    6. Dasgupta S,
    7. Ravi R,
    8. Guerrero-Preston R and
    9. Trink B
    : Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br J Cancer 105(9): 1295-1301, 2011. PMID: 21979421. DOI: 10.1038/bjc.2011.386
    OpenUrlCrossRefPubMed
  16. ↵
    1. Partecke LI,
    2. Evert K,
    3. Haugk J,
    4. Doering F,
    5. Normann L,
    6. Diedrich S,
    7. Weiss FU,
    8. Evert M,
    9. Huebner NO,
    10. Guenther C,
    11. Heidecke CD,
    12. Kramer A,
    13. Bussiahn R,
    14. Weltmann KD,
    15. Pati O,
    16. Bender C and
    17. von Bernstorff W
    : Tissue tolerable plasma (TTP) induces apoptosis in pancreatic cancer cells in vitro and in vivo. BMC Cancer 12: 473, 2012. PMID: 23066891. DOI: 10.1186/1471-2407-12-473
    OpenUrlCrossRefPubMed
    1. Vaquero J,
    2. Judée F,
    3. Vallette M,
    4. Decauchy H,
    5. Arbelaiz A,
    6. Aoudjehane L,
    7. Scatton O,
    8. Gonzalez-Sanchez E,
    9. Merabtene F,
    10. Augustin J,
    11. Housset C,
    12. Dufour T and
    13. Fouassier L
    : Cold-atmospheric plasma induces tumor cell death in preclinical in vivo and in vitro models of human cholangiocarcinoma. Cancers (Basel) 12(5): 1280, 2020. PMID: 32438553. DOI: 10.3390/cancers12051280
    OpenUrlCrossRefPubMed
    1. Utsumi F,
    2. Kajiyama H,
    3. Nakamura K,
    4. Tanaka H,
    5. Mizuno M,
    6. Ishikawa K,
    7. Kondo H,
    8. Kano H,
    9. Hori M and
    10. Kikkawa F
    : Effect of indirect nonequilibrium atmospheric pressure plasma on anti-proliferative activity against chronic chemo-resistant ovarian cancer cells in vitro and in vivo. PLoS One 8(12): e81576, 2013. PMID: 24367486. DOI: 10.1371/journal.pone.0081576
    OpenUrlCrossRefPubMed
    1. Alimohammadi M,
    2. Golpur M,
    3. Sohbatzadeh F,
    4. Hadavi S,
    5. Bekeschus S,
    6. Niaki HA,
    7. Valadan R and
    8. Rafiei A
    : Cold atmospheric plasma is a potent tool to improve chemotherapy in melanoma in vitro and in vivo. Biomolecules 10(7): 1011, 2020. PMID: 32650505. DOI: 10.3390/biom10071011
    OpenUrlCrossRefPubMed
  17. ↵
    1. Koensgen D,
    2. Besic I,
    3. Gümbel D,
    4. Kaul A,
    5. Weiss M,
    6. Diesing K,
    7. Kramer A,
    8. Bekeschus S,
    9. Mustea A and
    10. Stope MB
    : Cold atmospheric plasma (CAP) and CAP-stimulated cell culture media suppress ovarian cancer cell growth - a putative treatment option in ovarian cancer therapy. Anticancer Res 37(12): 6739-6744, 2017. PMID: 29187451. DOI: 10.21873/anticanres.12133
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Privat-Maldonado A,
    2. Gorbanev Y,
    3. Dewilde S,
    4. Smits E and
    5. Bogaerts A
    : Reduction of human glioblastoma spheroids using cold atmospheric plasma: the combined effect of short- and long-lived reactive species. Cancers (Basel) 10(11): 394, 2018. PMID: 30360539. DOI: 10.3390/cancers10110394
    OpenUrlCrossRefPubMed
    1. Cheng X,
    2. Sherman J,
    3. Murphy W,
    4. Ratovitski E,
    5. Canady J and
    6. Keidar M
    : The effect of tuning cold plasma composition on glioblastoma cell viability. PLoS One 9(5): e98652, 2014. PMID: 24878760. DOI: 10.1371/journal.pone.0098652
    OpenUrlCrossRefPubMed
  19. ↵
    1. Daeschlein G,
    2. Hillmann A,
    3. Gümbel D,
    4. Sicher C,
    5. von Podewils S,
    6. Stope MB and
    7. Jünger M
    : Enhanced anticancer efficacy by drug chemotherapy and cold atmospheric plasma against melanoma and glioblastoma cell lines in vitro. IEEE Transact Radiation Plasma Med Sci 2: 153-159, 2018. DOI: 10.1109/TRPMS.2018.2789659
    OpenUrlCrossRef
  20. ↵
    1. Chen Z,
    2. Simonyan H,
    3. Cheng X,
    4. Gjika E,
    5. Lin L,
    6. Canady J,
    7. Sherman JH,
    8. Young C and
    9. Keidar M
    : A novel micro cold atmospheric plasma device for glioblastoma both in vitro and in vivo. Cancers (Basel) 9(6): 61, 2017. PMID: 28555065. DOI: 10.3390/cancers9060061
    OpenUrlCrossRefPubMed
  21. ↵
    1. Köritzer J,
    2. Boxhammer V,
    3. Schäfer A,
    4. Shimizu T,
    5. Klämpfl TG,
    6. Li YF,
    7. Welz C,
    8. Schwenk-Zieger S,
    9. Morfill GE,
    10. Zimmermann JL and
    11. Schlegel J
    : Restoration of sensitivity in chemo-resistant glioma cells by cold atmospheric plasma. PLoS One 8(5): e64498, 2013. PMID: 23704990. DOI: 10.1371/journal.pone.0064498
    OpenUrlCrossRefPubMed
  22. ↵
    1. Gjika E,
    2. Pal-Ghosh S,
    3. Kirschner ME,
    4. Lin L,
    5. Sherman JH,
    6. Stepp MA and
    7. Keidar M
    : Combination therapy of cold atmospheric plasma (CAP) with temozolomide in the treatment of U87MG glioblastoma cells. Sci Rep 10(1): 16495, 2020. PMID: 33020527. DOI: 10.1038/s41598-020-73457-7
    OpenUrlCrossRefPubMed
  23. ↵
    1. Bekeschus S,
    2. Ispirjan M,
    3. Freund E,
    4. Kinnen F,
    5. Moritz J,
    6. Saadati F,
    7. Eckroth J,
    8. Singer D,
    9. Stope MB,
    10. Wende K,
    11. Ritter CA,
    12. Schroeder HWS and
    13. Marx S
    : Gas plasma exposure of glioblastoma is cytotoxic and immunomodulatory in patient-derived GBM tissue. Cancers (Basel) 14(3): 813, 2022. PMID: 35159079. DOI: 10.3390/cancers14030813
    OpenUrlCrossRefPubMed
  24. ↵
    1. Chautard E,
    2. Ouédraogo ZG,
    3. Biau J and
    4. Verrelle P
    : Role of Akt in human malignant glioma: from oncogenesis to tumor aggressiveness. J Neurooncol 117(2): 205-215, 2014. PMID: 24477623. DOI: 10.1007/s11060-014-1382-9
    OpenUrlCrossRefPubMed
  25. ↵
    1. Manning BD and
    2. Toker A
    : AKT/PKB signaling: navigating the network. Cell 169(3): 381-405, 2017. PMID: 28431241. DOI: 10.1016/j.cell.2017.04.001
    OpenUrlCrossRefPubMed
  26. ↵
    1. Maehama T and
    2. Dixon JE
    : The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem 273(22): 13375-13378, 1998. PMID: 9593664. DOI: 10.1074/jbc.273.22.13375
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Zinda MJ,
    2. Vlahos CJ and
    3. Lai MT
    : Ceramide induces the dephosphorylation and inhibition of constitutively activated Akt in PTEN negative U87mg cells. Biochem Biophys Res Commun 280(4): 1107-1115, 2001. PMID: 11162641. DOI: 10.1006/bbrc.2000.4248
    OpenUrlCrossRefPubMed
  28. ↵
    1. Koundouros N and
    2. Poulogiannis G
    : Phosphoinositide 3-Kinase/Akt signaling and redox metabolism in cancer. Front Oncol 8: 160, 2018. PMID: 29868481. DOI: 10.3389/fonc.2018.00160
    OpenUrlCrossRefPubMed
  29. ↵
    1. Edwards LA,
    2. Thiessen B,
    3. Dragowska WH,
    4. Daynard T,
    5. Bally MB and
    6. Dedhar S
    : Inhibition of ILK in PTEN-mutant human glioblastomas inhibits PKB/Akt activation, induces apoptosis, and delays tumor growth. Oncogene 24(22): 3596-3605, 2005. PMID: 15782140. DOI: 10.1038/sj.onc.1208427
    OpenUrlCrossRefPubMed
  30. ↵
    1. Kim SJ,
    2. Jung HJ and
    3. Lim CJ
    : Reactive oxygen species-dependent down-regulation of tumor suppressor genes PTEN, USP28, DRAM, TIGAR, and CYLD under oxidative stress. Biochem Genet 51(11-12): 901-915, 2013. PMID: 23832602. DOI: 10.1007/s10528-013-9616-7
    OpenUrlCrossRefPubMed
  31. ↵
    1. Wen C,
    2. Wang H,
    3. Wu X,
    4. He L,
    5. Zhou Q,
    6. Wang F,
    7. Chen S,
    8. Huang L,
    9. Chen J,
    10. Wang H,
    11. Ye W,
    12. Li W,
    13. Yang X,
    14. Liu H and
    15. Peng J
    : ROS-mediated inactivation of the PI3K/AKT pathway is involved in the antigastric cancer effects of thioredoxin reductase-1 inhibitor chaetocin. Cell Death Dis 10(11): 809, 2019. PMID: 31649256. DOI: 10.1038/s41419-019-2035-x
    OpenUrlCrossRefPubMed
  32. ↵
    1. Lopez-Gines C,
    2. Gil-Benso R,
    3. Benito R,
    4. Mata M,
    5. Pereda J,
    6. Sastre J,
    7. Roldan P,
    8. Gonzalez-Darder J and
    9. Cerdá-Nicolás M
    : The activation of ERK1/2 MAP kinases in glioblastoma pathobiology and its relationship with EGFR amplification. Neuropathology 28(5): 507-515, 2008. PMID: 18410277. DOI: 10.1111/j.1440-1789.2008.00911.x
    OpenUrlCrossRefPubMed
  33. ↵
    1. Zimmermann T,
    2. Gebhardt LA,
    3. Kreiss L,
    4. Schneider C,
    5. Arndt S,
    6. Karrer S,
    7. Friedrich O,
    8. Fischer MJM and
    9. Bosserhoff AK
    : Acidified nitrite contributes to the antitumor effect of cold atmospheric plasma on melanoma cells. Int J Mol Sci 22(7): 3757, 2021. PMID: 33916572. DOI: 10.3390/ijms22073757
    OpenUrlCrossRefPubMed
  34. ↵
    1. Nakai N,
    2. Fujita R,
    3. Kawano F,
    4. Takahashi K,
    5. Ohira T,
    6. Shibaguchi T,
    7. Nakata K and
    8. Ohira Y
    : Retardation of C2C12 myoblast cell proliferation by exposure to low-temperature atmospheric plasma. J Physiol Sci 64(5): 365-375, 2014. PMID: 25034108. DOI: 10.1007/s12576-014-0328-5
    OpenUrlCrossRefPubMed
  35. ↵
    1. Arndt S,
    2. Wacker E,
    3. Li YF,
    4. Shimizu T,
    5. Thomas HM,
    6. Morfill GE,
    7. Karrer S,
    8. Zimmermann JL and
    9. Bosserhoff AK
    : Cold atmospheric plasma, a new strategy to induce senescence in melanoma cells. Exp Dermatol 22(4): 284-289, 2013. PMID: 23528215. DOI: 10.1111/exd.12127
    OpenUrlCrossRefPubMed
  36. ↵
    1. Pearson G,
    2. Robinson F,
    3. Beers Gibson T,
    4. Xu BE,
    5. Karandikar M,
    6. Berman K and
    7. Cobb MH
    : Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 22(2): 153-183, 2001. PMID: 11294822. DOI: 10.1210/edrv.22.2.0428
    OpenUrlCrossRefPubMed
  37. ↵
    1. McCubrey JA,
    2. Steelman LS,
    3. Chappell WH,
    4. Abrams SL,
    5. Wong EW,
    6. Chang F,
    7. Lehmann B,
    8. Terrian DM,
    9. Milella M,
    10. Tafuri A,
    11. Stivala F,
    12. Libra M,
    13. Basecke J,
    14. Evangelisti C,
    15. Martelli AM and
    16. Franklin RA
    : Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 1773(8): 1263-1284, 2007. PMID: 17126425. DOI: 10.1016/j.bbamcr.2006.10.001
    OpenUrlCrossRefPubMed
  38. ↵
    1. Chetram MA and
    2. Hinton CV
    : PTEN regulation of ERK1/2 signaling in cancer. J Recept Signal Transduct Res 32(4): 190-195, 2012. PMID: 22737980. DOI: 10.3109/10799893.2012.695798
    OpenUrlCrossRefPubMed
  39. ↵
    1. Whitesell L and
    2. Lindquist SL
    : HSP90 and the chaperoning of cancer. Nat Rev Cancer 5(10): 761-772, 2005. PMID: 16175177. DOI: 10.1038/nrc1716
    OpenUrlCrossRefPubMed
  40. ↵
    1. Siegelin MD,
    2. Plescia J,
    3. Raskett CM,
    4. Gilbert CA,
    5. Ross AH and
    6. Altieri DC
    : Global targeting of subcellular heat shock protein-90 networks for therapy of glioblastoma. Mol Cancer Ther 9(6): 1638-1646, 2010. PMID: 20501802. DOI: 10.1158/1535-7163.MCT-10-0097
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. Li J and
    2. Buchner J
    : Structure, function and regulation of the hsp90 machinery. Biomed J 36(3): 106-117, 2013. PMID: 23806880. DOI: 10.4103/2319-4170.113230
    OpenUrlCrossRefPubMed
  42. ↵
    1. Stope MB,
    2. Koensgen D,
    3. Burchardt M,
    4. Concin N,
    5. Zygmunt M and
    6. Mustea A
    : Jump in the fire—heat shock proteins and their impact on ovarian cancer therapy. Crit Rev Oncol Hematol 97: 152-156, 2016. PMID: 26318096. DOI: 10.1016/j.critrevonc.2015.08.008
    OpenUrlCrossRefPubMed
  43. ↵
    1. Hermisson M,
    2. Strik H,
    3. Rieger J,
    4. Dichgans J,
    5. Meyermann R and
    6. Weller M
    : Expression and functional activity of heat shock proteins in human glioblastoma multiforme. Neurology 54(6): 1357-1365, 2000. PMID: 10746610. DOI: 10.1212/wnl.54.6.1357
    OpenUrlCrossRefPubMed
  44. ↵
    1. Sauvageot CM,
    2. Weatherbee JL,
    3. Kesari S,
    4. Winters SE,
    5. Barnes J,
    6. Dellagatta J,
    7. Ramakrishna NR,
    8. Stiles CD,
    9. Kung AL,
    10. Kieran MW and
    11. Wen PY
    : Efficacy of the HSP90 inhibitor 17-AAG in human glioma cell lines and tumorigenic glioma stem cells. Neuro Oncol 11(2): 109-121, 2009. PMID: 18682579. DOI: 10.1215/15228517-2008-060
    OpenUrlCrossRefPubMed
  45. ↵
    1. Chen H,
    2. Gong Y,
    3. Ma Y,
    4. Thompson RC,
    5. Wang J,
    6. Cheng Z and
    7. Xue L
    : A brain-penetrating Hsp90 inhibitor NXD30001 inhibits glioblastoma as a monotherapy or in combination with radiation. Front Pharmacol 11: 974, 2020. PMID: 32695001. DOI: 10.3389/fphar.2020.00974
    OpenUrlCrossRefPubMed
  46. ↵
    1. Beck R,
    2. Verrax J,
    3. Gonze T,
    4. Zappone M,
    5. Pedrosa RC,
    6. Taper H,
    7. Feron O and
    8. Calderon PB
    : Hsp90 cleavage by an oxidative stress leads to its client proteins degradation and cancer cell death. Biochem Pharmacol 77(3): 375-383, 2009. PMID: 19014912. DOI: 10.1016/j.bcp.2008.10.019
    OpenUrlCrossRefPubMed
  47. ↵
    1. Bekeschus S,
    2. Lippert M,
    3. Diepold K,
    4. Chiosis G,
    5. Seufferlein T and
    6. Azoitei N
    : Physical plasma-triggered ROS induces tumor cell death upon cleavage of HSP90 chaperone. Sci Rep 9(1): 4112, 2019. PMID: 30858416. DOI: 10.1038/s41598-019-38580-0
    OpenUrlCrossRefPubMed
  48. ↵
    1. Weiss M,
    2. Gümbel D,
    3. Hanschmann EM,
    4. Mandelkow R,
    5. Gelbrich N,
    6. Zimmermann U,
    7. Walther R,
    8. Ekkernkamp A,
    9. Sckell A,
    10. Kramer A,
    11. Burchardt M,
    12. Lillig CH and
    13. Stope MB
    : Cold atmospheric plasma treatment induces anti-proliferative effects in prostate cancer cells by redox and apoptotic signaling pathways. PLoS One 10(7): e0130350, 2015. PMID: 26132846. DOI: 10.1371/journal.pone.0130350
    OpenUrlCrossRefPubMed
  49. ↵
    1. Feil L,
    2. Koch A,
    3. Utz R,
    4. Ackermann M,
    5. Barz J,
    6. Stope M,
    7. Krämer B,
    8. Wallwiener D,
    9. Brucker SY and
    10. Weiss M
    : Cancer-selective treatment of cancerous and non-cancerous human cervical cell models by a non-thermally operated electrosurgical argon plasma device. Cancers (Basel) 12(4): 1037, 2020. PMID: 32340164. DOI: 10.3390/cancers12041037
    OpenUrlCrossRefPubMed
  50. ↵
    1. Jung YS,
    2. Qian Y and
    3. Chen X
    : Examination of the expanding pathways for the regulation of p21 expression and activity. Cell Signal 22(7): 1003-1012, 2010. PMID: 20100570. DOI: 10.1016/j.cellsig.2010.01.013
    OpenUrlCrossRefPubMed
  51. ↵
    1. Moldovan GL,
    2. Pfander B and
    3. Jentsch S
    : PCNA, the maestro of the replication fork. Cell 129(4): 665-679, 2007. PMID: 17512402. DOI: 10.1016/j.cell.2007.05.003
    OpenUrlCrossRefPubMed
  52. ↵
    1. Schneider C,
    2. Arndt S,
    3. Zimmermann JL,
    4. Li Y,
    5. Karrer S and
    6. Bosserhoff AK
    : Cold atmospheric plasma treatment inhibits growth in colorectal cancer cells. Biol Chem 400(1): 111-122, 2018. PMID: 29908123. DOI: 10.1515/hsz-2018-0193
    OpenUrlCrossRefPubMed
  53. ↵
    1. Eggers B,
    2. Stope MB,
    3. Marciniak J,
    4. Götz W,
    5. Mustea A,
    6. Deschner J,
    7. Nokhbehsaim M and
    8. Kramer FJ
    : 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(4): 889, 2022. PMID: 35453639. DOI: 10.3390/biomedicines10040889
    OpenUrlCrossRefPubMed
  54. ↵
    1. Eggers B,
    2. Marciniak J,
    3. Memmert S,
    4. Kramer FJ,
    5. Deschner J and
    6. 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 108(4): 607-616, 2020. PMID: 32030565. DOI: 10.1007/s10266-020-00487-y
    OpenUrlCrossRefPubMed
  55. ↵
    1. Kordt M,
    2. Trautmann I,
    3. Schlie C,
    4. Lindner T,
    5. Stenzel J,
    6. Schildt A,
    7. Boeckmann L,
    8. Bekeschus S,
    9. Kurth J,
    10. Krause BJ,
    11. Vollmar B and
    12. Grambow E
    : Multimodal imaging techniques to evaluate the anticancer effect of cold atmospheric pressure plasma. Cancers (Basel) 13(10): 2483, 2021. PMID: 34069689. DOI: 10.3390/cancers13102483
    OpenUrlCrossRefPubMed
  56. ↵
    1. Han C,
    2. Shen JK,
    3. Hornicek FJ,
    4. Kan Q and
    5. Duan Z
    : Regulation of microRNA-1 (miR-1) expression in human cancer. Biochim Biophys Acta Gene Regul Mech 1860(2): 227-232, 2017. PMID: 27923712. DOI: 10.1016/j.bbagrm.2016.12.004
    OpenUrlCrossRefPubMed
  57. ↵
    1. Bronisz A,
    2. Wang Y,
    3. Nowicki MO,
    4. Peruzzi P,
    5. Ansari K,
    6. Ogawa D,
    7. Balaj L,
    8. De Rienzo G,
    9. Mineo M,
    10. Nakano I,
    11. Ostrowski MC,
    12. Hochberg F,
    13. Weissleder R,
    14. Lawler SE,
    15. Chiocca EA and
    16. Godlewski J
    : Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res 74(3): 738-750, 2014. PMID: 24310399. DOI: 10.1158/0008-5472.CAN-13-2650
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Weiss M,
    2. Brandenburg LO,
    3. Burchardt M and
    4. Stope MB
    : MicroRNA-1 properties in cancer regulatory networks and tumor biology. Crit Rev Oncol Hematol 104: 71-77, 2016. PMID: 27286699. DOI: 10.1016/j.critrevonc.2016.05.014
    OpenUrlCrossRefPubMed
  59. ↵
    1. Soni V,
    2. Adhikari M,
    3. Simonyan H,
    4. Lin L,
    5. Sherman JH,
    6. Young CN and
    7. Keidar M
    : In vitro and in vivo enhancement of temozolomide effect in human glioblastoma by non-invasive application of cold atmospheric plasma. Cancers (Basel) 13(17): 4485, 2021. PMID: 34503293. DOI: 10.3390/cancers13174485
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 43 (1)
Anticancer Research
Vol. 43, Issue 1
January 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Devitalization of Glioblastoma Cancer Cells by Non-invasive Physical Plasma: Modulation of Proliferative Signalling Cascades
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 18 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Devitalization of Glioblastoma Cancer Cells by Non-invasive Physical Plasma: Modulation of Proliferative Signalling Cascades
SEBASTIAN LEHMANN, SANDRA BIEN-MÖLLER, SASCHA MARX, SANDER BEKESCHUS, HENRY W.S. SCHROEDER, ALEXANDER MUSTEA, MATTHIAS B. STOPE
Anticancer Research Jan 2023, 43 (1) 7-18; DOI: 10.21873/anticanres.16128

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Devitalization of Glioblastoma Cancer Cells by Non-invasive Physical Plasma: Modulation of Proliferative Signalling Cascades
SEBASTIAN LEHMANN, SANDRA BIEN-MÖLLER, SASCHA MARX, SANDER BEKESCHUS, HENRY W.S. SCHROEDER, ALEXANDER MUSTEA, MATTHIAS B. STOPE
Anticancer Research Jan 2023, 43 (1) 7-18; DOI: 10.21873/anticanres.16128
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • 5-Azacytidine (5-aza) Induces p53-associated Cell Death Through Inhibition of DNA Methyltransferase Activity in Hep3B and HT-29 Cells
  • Prognostic Value of WNT1, NOTCH1, PDGFRβ, and CXCR4 in Oral Squamous Cell Carcinoma
  • Hypoxia-adapted Multiple Myeloma Stem Cells Resist γδ-T-Cell-mediated Killing by Modulating the Mevalonate Pathway
Show more Experimental Studies

Similar Articles

Keywords

  • Cold atmospheric plasma
  • cold atmosphere pressure plasma
  • tissue tolerable plasma
  • microRNA-1
  • AKT1
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire