Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

1800MHz Microwave Induces p53 and p53-Mediated Caspase-3 Activation Leading to Cell Apoptosis In Vitro

  • Fuqiang Xing,

    Affiliations SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China, Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China

  • Qiuqiang Zhan,

    Affiliation SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China

  • Yiduo He,

    Affiliation Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China

  • Jiesheng Cui,

    Affiliation Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China

  • Sailing He ,

    sailing@jorcep.org (SH); wanggy@sustc.edu.cn (GW)

    Affiliations SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China, Department of Electromagnetic Engineering, Royal Institute of Technology (KTH), 10044 Stockholm, Sweden

  • Guanyu Wang

    sailing@jorcep.org (SH); wanggy@sustc.edu.cn (GW)

    Affiliation Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China

Abstract

Recent studies have reported that exposure of mammalian cells to microwave radiation may have adverse effects such as induction of cell apoptosis. However, the molecular mechanisms underlying microwave induced mammalian cell apoptosis are not fully understood. Here, we report a novel mechanism: exposure to 1800MHz microwave radiation induces p53-dependent cell apoptosis through cytochrome c-mediated caspase-3 activation pathway. We first measured intensity of microwave radiation from several electronic devices with an irradiation detector. Mouse NIH/3T3 and human U-87 MG cells were then used as receivers of 1800MHz electromagnetic radiation (EMR) at a power density of 1209 mW/m2. Following EMR exposure, cells were analyzed for viability, intracellular reactive oxygen species (ROS) generation, DNA damage, p53 expression, and caspase-3 activity. Our analysis revealed that EMR exposure significantly decreased viability of NIH/3T3 and U-87 MG cells, and increased caspase-3 activity. ROS burst was observed at 6 h and 48 h in NIH/3T3 cells, while at 3 h in U-87 MG cells. Hoechst 33258 staining and in situ TUNEL assay detected that EMR exposure increased DNA damage, which was significantly restrained in the presence of N-acetyl-L-cysteine (NAC, an antioxidant). Moreover, EMR exposure increased the levels of p53 protein and p53 target gene expression, promoted cytochrome c release from mitochondrion, and increased caspase-3 activity. These events were inhibited by pretreatment with NAC, pifithrin-α (a p53 inhibitor) and caspase inhibitor. Collectively, our findings demonstrate, for the first time, that 1800MHz EMR induces apoptosis-related events such as ROS burst and more oxidative DNA damage, which in turn promote p53-dependent caspase-3 activation through release of cytochrome c from mitochondrion. These findings thus provide new insights into physiological mechanisms underlying microwave-induced cell apoptosis.

Introduction

Human exposure to electromagnetic radiation (EMR) has increased dramatically in recent years, due to widespread use of various electronic devices, especially mobile phones. Devices that generate electromagnetic fields include radio or radar station transmitters, power transmission lines, high frequency welders, microwave ovens, and so on. Studies on the biological effects of EMR increase dramatically in recent years, as widespread uses of mobile phones have caused increasing concerns and debates regarding their implications to human health [1, 2]. Although it is still controversial about the risk to human health from EMR exposure, the International Agency for Research on Cancer (IARC) has evaluated human cancer risks from EMR exposure and classified EMR as a possible carcinogen to humans (2B) [3, 4].

Apoptosis is characterized by a number of genetic and biochemical events, including decreased cell viability, chromatin condensation, DNA fragmentation, and caspase activation. The use of mobile phones exposes human organs to frequent EMR. Recent studies have revealed a possible connection between EMR and impaired cell functions [5, 6], including the demonstration of increased apoptosis in human and animal cells exposed to 1800MHz EMR [7, 8]. Although those studies have demonstrated that EMR can induce cell apoptosis, the underlying molecular mechanisms remain largely unknown.

It is known that the nervous system, in particular the brain, is sensitive to EMR and other environmental factors[9]. Previous works have demonstrated that microwave radiation induces neuron apoptosis via the classical mitochondria-dependent caspase-3 pathway [10]. In addition, embryonic stem cells including mouse embryonic NIH/3T3 cells have been reported to be more sensitive to microwave exposure than differentiated cells. Therefore, they have been used frequently in environmental genotoxicity testing [11, 12]. In the present study, we shall use mouse NIH/3T3 and human U-87 MG cells as our model systems.

It has been established that reactive oxygen species (ROS) can damage various cellular compartments, leading to DNA damage, protein oxidation, lipid peroxidation and apoptosis[1315]. ROS is constantly produced under normal or mildly stressful conditions; and the basal concentration of ROS is usually pro-proliferative. Under severe stresses, excessive ROS is produced, which can damage DNA and proteins. Previous studies suggested that EMR exposure may affect living cells by increasing the ROS level and causing oxidative stresses [1618]. The tumor suppressor protein p53 is a transcription factor that mediates numerous extrinsic or intrinsic challenges to the cell, playing pivotal roles such as cell cycle arrest, apoptosis induction and DNA repair [19]. Activation of p53 upregulates pro-apoptosis genes; and the consequential apoptosis effectively prevents the accumulation of abnormal cells[20, 21]. In the present study, we focused on the potential roles played by ROS in cell apoptosis mediated by p53 signaling pathway and caused by 1800MHz EMR.

To test our hypothesis that microwave radiation induces cell apoptosis and to identify its biological mechanisms, we first measured the power densities of various electronic devices, and then selected a suitable one for further study. We then subjected NIH/3T3 and U-87 MG cells to microwave radiation with different time duration to measure their corresponding apoptosis. These works also allowed us to select the effective time duration for further investigation of the mechanism. To ensure that microwave exposure had induced cell apoptosis, we checked several indicators of apoptosis, such as DNA damage, release of cytochrome c from mitochondria and decrease in cell viability. Furthermore, we measured p53 expressions and caspase-3 activity, in both NIH/3T3 and U-87 MG cells subjected to 1800MHz radiation.

Materials and Methods

Reagents and antibodies

2’,7’-Dichlorodihydrofluorescin diacetate (DCFH-DA) and MitoSOX Red were purchased from Invitrogen (Carlsbad, California). The TdT-mediated X-dUTP nick end labeling (TUNEL) assay kit was purchased from Roche (Roche Molecular Biochemicals,Germany). Ac-DEVD-CHO, Z-VAD-FMKand the caspase-3 activity kit were purchased from Beyotime Institute of Biotechnology (Haimen, China). Hoechst 33258and N-Acetyl-L-cysteine (NAC) were obtained from Sigma (St. Louis, Missouri). Cell Counting Kit-8 (CCK-8) and pifithrin-α (PIF-α, p53 inhibitor) was purchased from Dojindo Laboratories (Kumamoto, Japan) and BioVision (Mountain View, CA, USA), respectively. Anti-p53, β-actin, anti-caspase-3, anti-cytochrome c antibodies, and all the secondary antibodies were obtained from Cell Signaling Technology (Beverly, MA).

Cell culture

The Mouse NIH/3T3 (Catalog No. GNM 6) and human U-87 MG (Catalog No. TCHu138) cell lines were purchased from Cellbank of the Chinese Academy of Sciences. Cells were cultured in Dulbecco’s modified Eagle’s medium (Gibco) supplemented with 10% Fetal bovine serum (HyClone), 100 U/ml penicillin and 100 μg/ml streptomycin (Gibco) at 5% CO2 and 37°C in a humidified incubator.

Measurements of microwave intensity

The power densities of various electronic devices were tested by an EMR detector according to a previously reported method[22]. Briefly, a 50MHz~3.5GHz X-Y-Z 3-dimensional EMR potential detector TES-92 was placed close to the surface of various electronic devices and the intensity of microwave was recorded (peak reading of XYZ mode).

The cell exposure system is composed of a microwave generator and transmitting antenna, the antenna is connected to a microwave generator via a flexible coaxial cable and enclosed in a conventional humidified incubator (5% CO2, 37°C). Petri dishes are positioned on the top of the antenna; cells were exposed to continuous microwave radiation at 1800MHz 1209 mW/m2 for different periods of time (0, 3, 6, 12, 24, 48 h).

Cell viability and apoptosis assay

Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan) was used in the current study to evaluate cell viability after various treatments. Optical density (OD450, the absorbance value at 450 nm) was directly read using a 96-well plate reader (imark, Bio-Rad, USA) from each microplate well. The value was normalized to that of the untreated cells from the same plate.

In situ BrdU-Red DNA Fragmentation (TUNEL) assay kit was used to detect DNA fragment. EMR treated cells were washed twice in PBS and added 50μl/well TUNEL reaction mixture, and were then incubated in a humidified atmosphere for 60 min at 37°C in the dark. Hoechst 33258 staining was performed following the previous method [23]. A stock solution of Hoechst 33258 was prepared at a concentration of 1 mg/ml in distilled water and stored at 4°C in the dark for use. EMR treated cells were washed twice in cold PBS and added Hoechst 33258 (final concentration: 10 μg/ml), and were then incubated at room temperature for 30 min in the dark. Cells whose nuclei exhibiting bright fluorescence, fragmented morphology and typical phenomena of nuclear condensation, were counted as apoptotic cells. Cells exhibiting diffusely fluorescent nuclei were classified as alive and viable. Fluorescence images of the cells were examined with a × 20 objective under a fluorescence microscope (Olympus IX71, Olympus Optical Co. Ltd, Tokyo, Japan).

Detection of intracellular ROS generation

Assessment of intracellular ROS changes in cells during apoptosis was performed using fluorescence probes H2DCFDA staining. A cell density of 105/ml was placed in each 35-mm culture plate (Thermo Fisher Scientific). After exposure, cells were preloaded with 10 μM DCFH-DA in phenol red-free culture medium for 30 min, then cells were detached using 0.25% trypsin (Gibco) and transferred to a 4-way cuvette (Hongjun Optics Co., China). Following digestion, the intensity of fluorescence was analyzed by a fluorescence spectrophotometer (F-2500, Hitachi) with excitation at 488 nm and emission at 525 nm. Each experiment was repeated three times.

MitoSOX Red was used to detect mitochondrial ROS production, cells were seeded at a density of 105/ml in glass bottom culture dishes (NEST, 801001). After exposure for 6 hours, cells were preloaded with 5 μM MitoSOX Red in phenol red-free culture medium for 30 min. Then cells were visualized by using two-photon fluoresence microscopy (Olympus FV1000 MPE). The wavelength of femtosecond pulse laser for two-photon excitation was 780 nm.

Caspase-3 enzymatic activities

The activity of caspase-3 was determined using the caspase-3 activity kit, based on the ability of caspase-3 to change acetyl-Asp-Glu-Val-Asp p-nitroanilide (Ac-DEVD-pNA) into a yellow formazan product p-nitroaniline (pNA). Caspase inhibitor Z-VAD-FMK (100 μM) and caspase-3-like inhibitor Ac-DEVD-CHO (100 μM) were used according to the manufacturer's instructions (Beyotime, Haimen, China). Lysates were centrifuged at 12,000 g for 10 min, and protein concentrations were determined by Bradford protein assay. Cellular extracts (30 μg) were incubated in a 96-well microtitre plate with 20 ng Ac-DEVD-pNA for 4 hours at 37°C. OD405 values were read with a 96-well plate reader. An increase in OD405 indicated the activation of caspase-3.

Protein extraction and western blot analysis

Detection of protein expression by western blot analysis was performed as described previously [24]. After different exposure treatments, cells were washed twice in cold PBS and lysed on ice with ice-cold lysis buffer (50 mM Tris HCl pH 8.0, 150 mM NaCl, 1% TritonX-100, 100 μg/ml PMSF) for 1 hour. The lysates were centrifuged (12.000g, 5 min, 4°C), followed by protein concentration determination. Equivalent cellular protein (30 μg each lane) were subjected to SDS-Polyacrylamide Gel Electrophoresis (SDS-PAGE) on 12% gel and transferred to a nitrocellulose membrane (Bio-Rad). The membranes with proteins were blocked with 5% skim milk for 0.5 hour and then incubated with primary antibodies at 4°C overnight followed by incubation with fluorescent secondary antibodies for 2 hours at room temperature. Dilution of primary and secondary antibody was 1:1000. Finally, the membranes were scanned on Bio-Rad's ChemiDoc XRS+ system with Image Lab image acquisition and analysis software. The intensity of the western blot signals was quantitated using ImageJ software, and the densitometry analyses are normalized, presented as the ratio protein/β-actin protein.

Real-time reverse transcription-PCR

Total RNA was isolated by standard trizol protocol and then converted into cDNA. The cDNA was amplified by real-time reverse transcription PCR (RT-PCR) using specific primers to quantify the expression of Bax, PUMA and GLS2, which are all p53 target genes. The primer sequences are presented in S1 Table. All qPCR reactions were performed using the CFX96 real time PCR detection system (Bio-Rad) with the following thermal cycle conditions: 95°C for 2 min, 39 cycles of (95°C for 5s, 60°C for 30 s), followed by a melt curve from 65°C to 95°C with an increment of 0.5°C per cycle. The level of inner loading controls were used to normalize expression of p53 target genes: GAPDH for the detection of Bax and PUMA expression in mouse NIH/3T3 cells; 36B4 (which encodes a ribosomal protein) for the detection of GLS2 and Bax expression in human U-87 MG cells.

Statistical analysis

Results were expressed as the mean-together with standard deviation (SD). Statistical analysis was performed with Student’s paired t-test using the statistical software SPSS, version10.0 (SPSS, Chicago). Differences were considered statistically significant at P<0.05 throughout the work. All charts were drawn using the software Origin, version 8.0.

Results

Sorting out suitable power density of antenna for cell exposure system

In consideration of the fact that power density of electronic devices used in our daily life, especially for cell phones, are constantly changing, we set up a fixed exposure system to assess the effects of EMR on cell physiology (Fig 1A and 1B). With the assistance of an EMR detector (the peak reading of XYZ mode), we tested the changes of microwave radiation power density near the surface of various electronic devices (Table 1 and S1 Fig).

thumbnail
Fig 1. The microwave exposure system.

(A) 1800MHz microwave generator. (B) Antenna in an incubator. The antenna was connected to a microwave generator via a flexible coaxial cable and enclosed in a humidified incubator. Petri dishes were positioned on the top of the antenna and cells in petri dishes were exposed to continuous microwave radiation at 1800MHz 1209 mW/m2.

https://doi.org/10.1371/journal.pone.0163935.g001

thumbnail
Table 1. Intensity test of microwave radiation for various electronic devices (peak reading XYZ mode).

https://doi.org/10.1371/journal.pone.0163935.t001

Radiation power density of cell phones (for both dialing and standby modes), microwave oven, personal computer and wireless router were monitored respectively (S1 Fig). We found that cell phone’s power density varies from 26 to 11330 mW/m2 during the dialing period; while it varies from 0.0035 to 7.425 mW/m2 during the standby period. Notably, the values of personal computer radiation were all below 10 mW/m2; wireless routers were a little higher; microwave ovens were much higher, reaching approximately 4500 mW/m2. We set 1209 mW/m2 as the peak reading of XYZ radiation of antenna at 1800MHz. As expected, the power density of antenna radiation used for this research was higher than the minimum value of cell phones (dialing time and standby time), personal computer and wireless router, but smaller than the maximum value of cell phones (dialing time) and microwave oven (working mode). Hence, these data indicate that 1209 mW/m2, the power density of antenna radiation at 1800MHz, can be used to detect the potential mechanisms of EMR induced cell apoptosis in this research.

1800MHz microwave induces caspase-3 activation and decrease in cell viability

To test whether 1800MHz microwave with power density strength at 1209 mW/m2 induces apoptosis in NIH/3T3 and U-87 MG cells, we determined the cell viability and caspase-3 activity after 1800MHz microwave irradiation. We found that the viability of NIH/3T3 cells decreased gradually from 100% to 55% over 48 hours during 1800MHz microwave irradiation (Fig 2A). Similarly, the viability of U-87 MG cells decreased slightly over 12 hours then rapidly from 24 hour to 48 hours (Fig 2B). We further measured caspase-3 activity after 1800MHz microwave irradiation. Compared with the sham group of NIH/3T3 cells, caspase-3 activity increased gradually over 24 hours, followed by an obvious escalation at 48 hours (Fig 2C). For U-87 MG cells, caspase-3 activity increased rapidly during the first three hours, and then maintained a gradual increase to 24 hours, then decreased (Fig 2D). In summary, these data indicate that 1800MHz microwave irradiation induces caspase-3 dependent apoptosis in NIH/3T3 and U-87 MG cells.

thumbnail
Fig 2. 1800 MHz microwave induces caspase-3 activation and decrease in cell viability.

(A, B) Levels of cell viability after 1800MHz microwave radiation for the indicated time in NIH/3T3 and U-87 MG cells. Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan) was used to evaluate cell viability after various treatments. (C, D) Levels of caspase-3 activity after 1800MHz microwave radiation for the indicated time in NIH/3T3and U-87 MG cells. After microwave radiation, cells were harvested, and caspase-3 activity was measured by the caspase-3 activity kit. MW, microwave. Data are the mean ± S.D. (n = 3; *P < 0.05 vs. sham group.)

https://doi.org/10.1371/journal.pone.0163935.g002

1800MHz microwave-induced oxidative DNA damage

To assess whether 1800MHz microwave irradiation can induce ROS generation in NIH/3T3 and U-87 MG cells, we measured intracellular ROS production by using a 2’,7’-dichlorofluorescin diacetate fluorescence probe. NIH/3T3 and U-87 MG cells were irradiated with 1800MHz microwave for different time intervals (0, 3, 6, 12, 24 and 48 hours). We found that, compared with the sham group, 1800MHz microwave irradiation led to fluorescence intensity changes fluctuating greatly over 48 hours in NIH/3T3 cells but only a rapid increase at 3 hours in U-87 MG cells (Fig 3A and 3B). We further detected whether 1800MHz microwave irradiation can induce mitochondrial ROS production by using MitoSOX Red probe on a two-photon laser scanning microscope system (Olympus FV1000 MPE). Our results showed that 1800MHz microwave exposure of NIH/3T3 and U-87 MG cells for 6 hours led to mitochondrial ROS production, which was inhibited by broad-spectrum antioxidant NAC (Fig 3C and 3D). Because nuclear condensation and DNA fragmentation are also hallmarks of cell apoptosis, we then determined whether 1800MHz microwave can induce nuclear condensation and DNA fragmentation by Hoechst 33258 staining and in situ TUNEL assay, respectively. Hoechst 33258 dye is a fluorescent nucleic acid stain for detecting nuclear condensation (Fig 4B and S2 Fig). TUNEL enables highly sensitive detection of apoptosis of single cells. Cells undergoing apoptosis were preferentially labeled by the TUNEL reaction (Fig 4A). To further confirm hypothesis that ROS generation is required for 1800MHz microwave induced DNA damage, NIH/3T3 and U-87 MG cells were pre-treated with the NAC and exposed continuously to EMR for different time intervals (0, 3, 6, 12, 24 and 48 hours). We found that the DNA damage increased NIH/3T3 cells more than two times than the increase of U-87 MG cells, after treatment with 1800MHz microwave (Fig 4C and 4D). These results indicate that 1800MHz microwave exposure of NIH/3T3 and U-87 MG cells cause DNA fragmentation and nuclear condensation. Interestingly, increases in DNA damage were significantly inhibited by NAC in NIH/3T3 cells, but only slightly in U-87 MG cells, supporting the hypothesis that ROS generation is required for DNA injury and cell apoptosis induced by 1800MHz microwave irradiation.

thumbnail
Fig 3. Exposure to 1800MHz microwave induces ROS production in NIH/3T3 and U-87 MG cells.

(A, B) Intracellular ROS production was measured by using DCFH-DA after 1800MHz microwave radiation for the indicated time in NIH/3T3 and U-87 MG cells. (C, D) detection of mitochondrial ROS production by using MitoSOX Red after 1800MHz microwave exposure for 6 hours in NIH/3T3 and U-87 MG cells. The imaging experiments were implemented on a two-photon laser scanning microscope system. DCF, 2’,7’-Dichlorodihydrofluorescin diacetate; mROS, mitochondrial reactive oxygen species; MW, microwave; NAC, N-acetyl-L-cysteine.Scale bars: 10 μm. Data represent mean ± S.D. (n = 3; *P < 0.05 vs. sham group.)

https://doi.org/10.1371/journal.pone.0163935.g003

thumbnail
Fig 4. Exposure to 1800MHz microwave induces oxidative DNA damage in NIH/3T3 and U-87 MG cells.

(A) In situ TUNEL detection of DNA fragmentation. (B) Hoechst 33258 staining assessment of nuclear condensation in NIH/3T3 and U-87 MG cells. Fluorescent images of DNA damaged cells were visualized under an Olympus fluorescent microscope (20×). (C, D) Quantitative analysis of DNA damages in NIH/3T3 and U87-MG cells stained by Hoechst 33258 shown in (B). MW, microwave; NAC, N-acetyl-L-cysteine. Data represent mean ± S.D. (n = 3; *P < 0.05 vs. sham group.)

https://doi.org/10.1371/journal.pone.0163935.g004

1800MHz microwave-induced ROS production upregulates p53 expression

We then explored the mechanisms of p53 mediated regulation of cell apoptosis induced by 1800MHz microwave. After the irradiation, cells were harvested for protein and mRNA extraction to detect the level of p53 changes and the expression of genes targeted by p53. Western blotting analysis showed that the level of p53 significantly increased upon 1800MHz microwave irradiation relative to the sham group. But this was suppressed by incubation the cells with antioxidant NAC and this suppression effect of p53 expression by NAC was more notable in NIH/3T3 than in U-87 MG cells (Fig 5A and 5B). Moreover, real-time RT-PCR showed that 1800MHz microwave irradiation enhanced expression of p53 target genes: Bax, PUMA and GLS2. These enhanced expressions were significantly inhibited by antioxidant NAC and p53 inhibitor PIF-α (Fig 5C–5F). Together, these results suggested that ROS is involved in 1800MHz microwave-induced upregulation of p53 in cell apoptosis.

thumbnail
Fig 5. ROS upregulates p53 expression during 1800MHz microwave-induced cell apoptosis.

(A, B) ROS induces p53 expression upon 1800MHz microwave radiation for 24 hours in NIH/3T3 and U-87 MG cells. Cells were pretreated with or without NAC and exposed to 1800MHz. The representative western blot assay of p53 exposed to 1800MHz microwave in the presence of NAC in mouse NIH/3T3 and human U-87 MG cells, respectively. (C, D) Detection of the expression of Bax and PUMA, respectively, in mouse NIH/3T3 cells. (E,F) Detection of the expression of GLS2 and Bax, respectively, in human U-87 MG cells. The level of loading controls was used to normalize the expression of p53 target genes: Bax, PUMA, and GLS2. MW, microwave; NAC, N-acetyl-L-cysteine; PIF-α, pifithrin-α. All the data in these figures are presented as mean ± S.D. (n = 3; *P < 0.05 vs. indicated group.)

https://doi.org/10.1371/journal.pone.0163935.g005

Involvement of ROS and p53 in cytochrome c-mediated caspase-3 activation during 1800MHz microwave-induced cell apoptosis

Since caspase-3 activation and cytochrome c release from mitochondrion are key events in cell apoptosis [25, 26], our results showed that in the presence of Ac-DEVD-CHO (caspase-3 inhibitor) or Z-VAD-FMK (broad-spectrum caspase inhibitor) prevented 1800MHz microwave-induced increase of caspase-3 activity and the consequential decrease of cell viability (Fig 6A and 6B). We also assessed whether or not cell death induced by ROS and p53 upregulation was through cytochrome c-mediated caspase-3 activation pathway. NAC or PIF-α were incubated with NIH/3T3 and U-87 MG cells, respectively. These cells were then subjected to cells subjecting to 1800MHz microwave irradiation. We found that incubation with NAC or PIF-α had strongly blocked the decrease of cell viability and the activation of caspase-3 induced by 1800MHz microwave in NIH/3T3 cells and U-87 MG cells, respectively (Fig 6A and 6B).

thumbnail
Fig 6. ROS and p53 involve in cytochrome c-mediated caspase-3 activation during 1800MHz microwave-induced cell apoptosis.

(A, B) Effects of NAC, PIF-α, Ac-DEVD-CHO and Z-VAD-FMK on cell viability and caspase-3 activity after 1800MHz microwave radiation for 24 hours in NIH/3T3 and U-87 MG cells. Cells were pretreated with treatments (NAC, PIF-α, Ac-DEVD-CHO and Z-VAD-FMK) for 20 min and then exposed to 1800MHz microwave or sham treatment for 24 hours. After microwave radiation, cells were harvested and levels of cell viability and caspase-3 activity were measured in NIH/3T3 and U-87 MG cells. Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan) was used to evaluate cell viability after various treatments. Caspase-3 activity was measured by the caspase-3 activity kit. (C, D) Western blotting analysis of caspase-3 activation and cytochrome c release from mitochondria to cytoplasm. Cells were pretreated with or without NAC and PIF-α for 20 min before exposure to 1800MHz microwave. After microwave radiation for 24 hours, cells were harvested and measured protein levels of caspase-3. The cytoplasm proteins were extracted using a mitochondria/cytoplasm fractionation kit followed by western blotting analysis of cytochrome c in cytoplasm. MW, microwave; NAC, N-acetyl-L-cysteine. PIF-α, pifithrin-α. All the data in these figures are presented as mean ± S.D. (n = 3; *P < 0.05 vs. control group; #P < 0.05 indicated group.)

https://doi.org/10.1371/journal.pone.0163935.g006

We then performed western blotting analysis to assess cytochrome c release from mitochondria to cytoplasm. Our results showed that NAC and PIF-α can inhibit the increase in protein levels of caspase-3 and cytochrome c in NIH/3T3 cells and U-87 MG cytoplasm (Fig 6C and 6D). Together, these data suggest that ROS and p53 are required for cytochrome c-mediated caspase-3 activation induced by 1800MHz microwave in NIH/3T3 and U-87 MG cells.

Discussion

Numerous studies have been performed to assess human health risks and biological consequences of exposure to microwave radiation, which generated a large bulk of data[2733]. These data however seem to be contradictory, probably due to different sensitivities of the biological systems to EMR exposure and different parameters of electromagnetic fields. Both factors influence their interactions between EMR and living cells. For instance, single or multiple radiofrequency radiation exposure did not elicit oxidative stress in human breast epithelial MCF10A cells [34]. However, other studies indicated that oxidative stress caused by exposure to Wi-Fi or mobile phone EMR may significantly affect female and male reproductive systems[35]. EMR can also affect gene expression and modulate a wide range of biological processes such as cell-cycle control, apoptosis and autophagy [33, 36, 37]. The present study provides evidences that exposure of NIH/3T3 and U-87 MG cells to EMR at 1800MHz, with a specific power density at approximately 1209 mW/m2, induced apoptosis in each cell being investigated (Figs 2, 4 and 6).

Previous studies demonstrated that 1800MHz microwave exposure can induce a dose-dependent or time-dependent ROS generation and DNA damage in fibroblasts cell line [13, 38]. In our experiments, we observed a fluctuating ROS generation in NIH/3T3 cells and transient increase at 3 hours after EMR in U-87 MG cells (Fig 3A and 3B), this may largely due to antioxidant enzymes including superoxide dismutase, glutathione peroxidase, and catalase working together to defend the cells against toxic ROS. Since 1800MHz microwave can also directly induce DNA damage which will further promote p53 activation[39], we speculate that ROS generation and DNA damage may have a synergistic effect in regulating 1800MHz microwave induced p53 activation and cell apoptosis. De et al. found that mobile phones enhance mitochondrial ROS generation and stimulates DNA fragmentation[6]. But it remains to be determined if ROS is responsible for DNA damage, as well as possible pathways through which ROS can affect apoptosis. Here, we observed that 1800MHz microwave irradiation induces ROS generation, which causes DNA damages in NIH/3T3 and U-87 MG cells (Fig 4). Additionally, since 1800MHz microwave irradiation-induced DNA damages were not totally blocked by NAC in our experiment, it is possible that 1800MHz microwave directly induces DNA damage. Furthermore, exposure to intensive electromagnetic fields generated by devices such as radar and mobile phone transmitters can lead to electrical shock and even thermal effects. Many current researchers are also interested in the possible "non-thermal" effects. Previous reports suggested that mobile phone radiation usually does not cause heating but modulates signal transduction pathways and induces cellular stress response [4042]. In the present study, the dishes and antenna were placed in a humidified incubator (37°C, 5% CO2, 95% humidity) to ensure constant environmental conditions for studying the mechanisms of cell apoptosis induced by microwave (Fig 1).

Some conditions in the present study, for example the 48h constant exposure to EMR, seem not to be physiologically relevant. This might be true ten years ago. But with the fast development of wireless and IT industries nowadays, more and more electronic products emerge and more and more people suffer from prolonged EMR due to using these products. For cell phone alone, people use it more frequently, relying on it for communication, education, and e-commerce; it is replacing the laptop. Some people keep the phone near the brain (under the pillow); or near the reproductive system (in a trouser pocket). It is thus common to have a nearly constant exposure to GSM 900/1800MHz cell phone signal in the “on” mode (exposed) or in the ‘‘stand-by” mode (sham) for a long time, approaching or even exceeding 48 hours. To mimic these physiological conditions, we have put NIH/3T3 (embryo fibroblast) and U-87 MG (brain glioblastoma) cells under either exposure or sham (stand-by) conditions for up to 48 hours. Due to facility restrictions, we have used continuous microwaves but not the more realistic pulsed microwaves, which represent one limitation of our work.

Although p53 protein has been implicated to play roles in a variety of cellular processes, its role in cell apoptosis induced by microwave irradiation is still unknown. Here, we found that p53 and p53 target gene induced by 1800MHz microwave irradiation was inhibited by NAC and p53 inhibitor PIF-α pretreatment, suggesting that ROS is involved in regulating the expression of p53 (Fig 5). Besides ROS, many other signals can also activate p53 [43]. Some studies have demonstrated that p53 is extremely sensitive to even low levels of DNA damage, thus promoting DNA repair directly by upregulating the expression of DNA repair genes [44]. In the present study, since 1800MHz microwave irradiation-induced p53 activation was partially blocked by NAC, it is reasonable to speculate that ROS burst and DNA damage together may be involved in upregulating p53 expression during 1800MHz microwave irradiation induced apoptosis in NIH/3T3 and U-87 MG cells. Hussain et al. found that p53-mediated apoptosis may activates redox-controlling genes, resulting in ROS production [45]. In the present study, we confirmed the hypothesis that an oxidative stress induced by 1800MHz microwave irradiation plays a vital role in regulating p53-dependent caspase-3 activation. Caspase-3 is cysteine proteases with well-established functions in promoting apoptosis[4648]. It is well known that caspase-3 can be activated by proapoptotic molecules such as cytochrome c released from mitochondria[49, 50]. Mitochondria are a major source of ROS, mainly at the level of complexes I and III of the respiratory chain [51]. In the present experiment, our results suggested that EMR-induced the release of cytochrome c and caspase-3 activation had been blocked by NAC or PIF-α (Fig 6), it is thus safe to say that 1800MHz microwave irradiation induces mitochondrial ROS and the release of cytoplasmic cytochrome c and in turn, activates caspase-3 in NIH/3T3 and U-87 MG cells.

In conclusion, we report here, for the first time, that exposure of NIH/3T3 and U-87 MG cells to 1800MHz microwave radiation induces ROS production and DNA damage, which in turn up-regulates p53-dependent caspase-3 activation through cytochrome c. These events all promote cell apoptosis (Fig 7). Despite these novel findings, it remains unclear how radiation is perceived and how the signal is rapidly transduced into the cells. It is thus important to address these challenges in the future.

thumbnail
Fig 7. Schematic representation of p53 activation of caspase-3 signaling pathway in 1800MHz microwave induced cell apoptosis.

1800MHz microwave activates p53 signaling pathway through enhancing ROS production and DNA damage, p53 further upregulates cytochrome c-mediated caspase-3 activation that leads to cell apoptosis. MW, microwave; NAC, N-acetyl-L-cysteine; ROS, reactive oxygen species.

https://doi.org/10.1371/journal.pone.0163935.g007

Supporting Information

S1 Fig. Determine the intensity of microwave radiation on the surface of various electronic devices.

The power densities of antenna,cell phone,microwave oven,personal computer andwireless router were measured by using a 50MHz~3.5GHz X-Y-Z 3-dimensional EMR potential detector TES-92 (peak reading of XYZ mode) on the surface of electronic devices.

https://doi.org/10.1371/journal.pone.0163935.s001

(TIF)

S2 Fig. Confocal microscopic detection of 1800MHz microwave-induced DNA damage.

Hoechst 33258 dye (a fluorescent nucleic acid stain) was used fordetecting nuclear condensation after NIH/3T3 and U-87 MG cells were irradiated with 1800MHz microwave for 48 hours. Cells were visualized by using two-photon fluoresence microscopy (Olympus FV1000 MPE). The wavelength of 730 nm was used for femtosecond pulse laser for two-photon excitation of Hoechst 33258 dye. Arrows indicate nuclei exhibiting bright fluorescence and the fragmented nuclear morphology.

https://doi.org/10.1371/journal.pone.0163935.s002

(TIF)

Acknowledgments

We sincerely thank Yuxiang Zhao and Xing Wan for assistance in the experiment, Julian Samuel Evans for useful discussion of the manuscript, and Brian Darville for assistance in checking the language.

Author Contributions

  1. Conceived and designed the experiments: FQX QQZ SLH GYW.
  2. Performed the experiments: FQX QQZ.
  3. Analyzed the data: FQX QQZ YDH JSC SLH GYW.
  4. Contributed reagents/materials/analysis tools: FQX GYW.
  5. Wrote the paper: FQX GYW.

References

  1. 1. Krause CM, Sillanmäki L, Koivisto M, Häggqvist A, Saarela C, Revonsuo A, et al. Effects of electromagnetic field emitted by cellular phones on the EEG during a memory task. Neuroreport. 2000;11(4):761–4. pmid:10757515
  2. 2. Huber R, Treyer V, Borbely A, Schuderer J, Gottselig J, Landolt HP, et al. Electromagnetic fields, such as those from mobile phones, alter regional cerebral blood flow and sleep and waking EEG. Journal of sleep research. 2002;11(4):289–95. pmid:12464096
  3. 3. Baan R, Grosse Y, Lauby-Secretan B, El Ghissassi F, Bouvard V, Benbrahim-Tallaa L, et al. Carcinogenicity of radiofrequency electromagnetic fields. The lancet oncology. 2011;12(7):624–6. pmid:21845765
  4. 4. Hardell L, Carlberg M, Söderqvist F, Hansson Mild K. Meta-analysis of long-term mobile phone use and the association with brain tumours. International journal of oncology. 2008;32(5):1097–103. pmid:18425337
  5. 5. Diem E, Schwarz C, Adlkofer F, Jahn O, Rüdiger H. Non-thermal DNA breakage by mobile-phone radiation (1800MHz) in human fibroblasts and in transformed GFSH-R17 rat granulosa cells in vitro. Mutation Research/Genetic Toxicology and Environmental Mutagenesis. 2005;583(2):178–83. pmid:15869902
  6. 6. De Iuliis GN, Newey RJ, King BV, Aitken RJ. Mobile phone radiation induces reactive oxygen species production and DNA damage in human spermatozoa in vitro. PloS one. 2009;4(7):e6446. pmid:19649291
  7. 7. Liu Y, Hong R, Yu Y, Weng E. [Effects of extremely low frequency electromagnetic fields on apoptosis and cell cycle of mouse brain and liver cells]. Zhonghua lao dong wei sheng zhi ye bing za zhi = Zhonghua laodong weisheng zhiyebing zazhi = Chinese journal of industrial hygiene and occupational diseases. 2003;21(5):339–41. pmid:14761394
  8. 8. Caraglia M, Marra M, Mancinelli F, D'ambrosio G, Massa R, Giordano A, et al. Electromagnetic fields at mobile phone frequency induce apoptosis and inactivation of the multi-chaperone complex in human epidermoid cancer cells. Journal of cellular physiology. 2005;204(2):539–48. pmid:15754340
  9. 9. Nikolova T, Czyz J, Rolletschek A, Blyszczuk P, Fuchs J, Jovtchev G, et al. Electromagnetic fields affect transcript levels of apoptosis-related genes in embryonic stem cell-derived neural progenitor cells. The FASEB journal. 2005;19(12):1686–8. pmid:16116041
  10. 10. Zuo H, Lin T, Wang D, Peng R, Wang S, Gao Y, et al. Neural Cell Apoptosis Induced by Microwave Exposure Through Mitochondria-dependent Caspase-3 Pathway. International journal of medical sciences. 2014;11(5):426. pmid:24688304
  11. 11. Hou Q, Wang M, Wu S, Ma X, An G, Liu H, et al. Oxidative changes and apoptosis induced by 1800-MHz electromagnetic radiation in NIH/3T3 cells. Electromagnetic biology and medicine. 2014;(0):1–8.
  12. 12. Yang L, Hao D, Wang M, Zeng Y, Wu S, Zeng Y. Cellular neoplastic transformation induced by 916 MHz microwave radiation. Cellular and molecular neurobiology. 2012;32(6):1039–46. pmid:22395787
  13. 13. Liu C, Duan W, Xu S, Chen C, He M, Zhang L, et al. Exposure to 1800MHz radiofrequency electromagnetic radiation induces oxidative DNA base damage in a mouse spermatocyte-derived cell line. Toxicology letters. 2013;218(1):2–9. pmid:23333639
  14. 14. Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, et al. Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. Journal of Biological Chemistry. 2003;278(10):8516–25. pmid:12496265
  15. 15. Simon H-U, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 2000;5(5):415–8. pmid:11256882
  16. 16. Zmyślony M, Politanski P, Rajkowska E, Szymczak W, Jajte J. Acute exposure to 930 MHz CW electromagnetic radiation in vitro affects reactive oxygen species level in rat lymphocytes treated by iron ions. Bioelectromagnetics. 2004;25(5):324–8. pmid:15197754
  17. 17. Lantow M, Lupke M, Frahm J, Mattsson M, Kuster N, Simko M. ROS release and Hsp70 expression after exposure to 1,800 MHz radiofrequency electromagnetic fields in primary human monocytes and lymphocytes. Radiation and environmental biophysics. 2006;45(1):55–62. pmid:16552570
  18. 18. Lantow M, Schuderer J, Hartwig C, Simko M. Free radical release and HSP70 expression in two human immune-relevant cell lines after exposure to 1800 MHz radiofrequency radiation. Radiation research. 2006;165(1):88–94. pmid:16392966
  19. 19. Vousden KH, Lane DP. p53 in health and disease. Nature reviews Molecular cell biology. 2007;8(4):275–83. pmid:17380161
  20. 20. Sheikh MS, Fornace AJ. Role of p53 family members in apoptosis. Journal of cellular physiology. 2000;182(2):171–81. pmid:10623880
  21. 21. Amaral JD, Xavier JM, Steer CJ, Rodrigues C. The role of p53 in apoptosis. Discovery medicine. 2010;9(45):145–52. pmid:20193641
  22. 22. Lin C-S, editor Low power 60 kHz electrodeless fluorescent lamp for indoor use. IPEC, 2010 Conference Proceedings; 2010: IEEE.
  23. 23. Xu L, Yenari MA, Steinberg GK, Giffard RG. Mild hypothermia reduces apoptosis of mouse neurons in vitro early in the cascade. Journal of Cerebral Blood Flow & Metabolism. 2002;22(1):21–8. pmid:11807390
  24. 24. Meng C, He Z, Xing D. Low-level laser therapy rescues dendrite atrophy via upregulating BDNF expression: implications for Alzheimer's disease. The Journal of Neuroscience. 2013;33(33):13505–17. pmid:23946409
  25. 25. Yang Y, Xing D, Zhou F, Chen Q. Mitochondrial autophagy protects against heat shock-induced apoptosis through reducing cytosolic cytochrome c release and downstream caspase-3 activation. Biochemical and biophysical research communications. 2010;395(2):190–5. pmid:20361931
  26. 26. Shi L, Chen J, Yang J, Pan T, Zhang S, Wang Z. MiR-21 protected human glioblastoma U87MG cells from chemotherapeutic drug temozolomide induced apoptosis by decreasing Bax/Bcl-2 ratio and caspase-3 activity. Brain research. 2010;1352:255–64. pmid:20633539
  27. 27. Kesari KK, Kumar S, Behari J. 900-MHz microwave radiation promotes oxidation in rat brain. Electromagnetic Biology and Medicine. 2011;30(4):219–34. pmid:22047460
  28. 28. Schwarz C, Kratochvil E, Pilger A, Kuster N, Adlkofer F, Rüdiger HW. Radiofrequency electromagnetic fields (UMTS, 1,950 MHz) induce genotoxic effects in vitro in human fibroblasts but not in lymphocytes. International archives of occupational and environmental health. 2008;81(6):755–67. pmid:18278508
  29. 29. Zhao T-Y, Zou S-P, Knapp PE. Exposure to cell phone radiation up-regulates apoptosis genes in primary cultures of neurons and astrocytes. Neuroscience letters. 2007;412(1):34–8. pmid:17187929
  30. 30. Schüz J, Steding-Jessen M, Hansen S, Stangerup S-E, Cayé-Thomasen P, Poulsen AH, et al. Long-term mobile phone use and the risk of vestibular schwannoma: a Danish nationwide cohort study. American journal of epidemiology. 2011;174(4):416–22. pmid:21712479
  31. 31. Markovà E, Hillert L, Malmgren L, Persson BR, Belyaev IY. Microwaves from GSM mobile telephones affect 53BP1 and γ-H2AX foci in human lymphocytes from hypersensitive and healthy persons. Environmental health perspectives. 2005:1172–7. pmid:16140623
  32. 32. Belyaev IY, Koch CB, Terenius O, Roxström-Lindquist K, Malmgren LO, H Sommer W, et al. Exposure of rat brain to 915 MHz GSM microwaves induces changes in gene expression but not double stranded DNA breaks or effects on chromatin conformation. Bioelectromagnetics. 2006;27(4):295–306. pmid:16511873
  33. 33. Buttiglione M, Roca L, Montemurno E, Vitiello F, Capozzi V, Cibelli G. Radiofrequency radiation (900 MHz) induces Egr-1 gene expression and affects cell-cycle control in human neuroblastoma cells. Journal of cellular physiology. 2007;213(3):759–67. pmid:17559061
  34. 34. Hong MN, Kim BC, Ko YG, Lee YS, Hong SC, Kim T, et al. Effects of 837 and 1950 MHz radiofrequency radiation exposure alone or combined on oxidative stress in MCF10A cells. Bioelectromagnetics. 2012;33(7):604–11. pmid:22549623
  35. 35. Nazıroğlu M, Yüksel M, Köse SA, Özkaya MO. Recent reports of Wi-Fi and mobile phone-induced radiation on oxidative stress and reproductive signaling pathways in females and males. The Journal of membrane biology. 2013;246(12):869–75. pmid:24105626
  36. 36. Marchesi N, Osera C, Fassina L, Amadio M, Angeletti F, Morini M, et al. Autophagy is modulated in human neuroblastoma cells through direct exposition to low frequency electromagnetic fields. Journal of cellular physiology. 2014;229(11):1776–86. pmid:24676932
  37. 37. Xu S, Zhou Z, Zhang L, Yu Z, Zhang W, Wang Y, et al. Exposure to 1800 MHz radiofrequency radiation induces oxidative damage to mitochondrial DNA in primary cultured neurons. Brain Research. 2010;1311:189–96. pmid:19879861
  38. 38. Ivancsits S, Diem E, Jahn O, Rüdiger HW. Intermittent extremely low frequency electromagnetic fields cause DNA damage in a dose-dependent way. International archives of occupational and environmental health. 2003;76(6):431–6. pmid:12802592
  39. 39. Takagi M, Absalon MJ, McLure KG, Kastan MB. Regulation of p53 translation and induction after DNA damage by ribosomal protein L26 and nucleolin. Cell. 2005;123(1):49–63. pmid:16213212
  40. 40. Leszczynski D, Joenväärä S, Reivinen J, Kuokka R. Non-thermal activation of the hsp27/p38MAPK stress pathway by mobile phone radiation in human endothelial cells: molecular mechanism for cancer-and blood-brain barrier-related effects. Differentiation. 2002;70(2):120–9. pmid:12076339
  41. 41. Di Carlo A, White N, Guo F, Garrett P, Litovitz T. Chronic electromagnetic field exposure decreases HSP70 levels and lowers cytoprotection. Journal of cellular biochemistry. 2002;84(3):447–54. pmid:11813250
  42. 42. Mashevich M, Folkman D, Kesar A, Barbul A, Korenstein R, Jerby E, et al. Exposure of human peripheral blood lymphocytes to electromagnetic fields associated with cellular phones leads to chromosomal instability. Bioelectromagnetics. 2003;24(2):82–90. pmid:12524674
  43. 43. Derdak Z, Lang CH, Villegas KA, Tong M, Mark NM, Suzanne M, et al. Activation of p53 enhances apoptosis and insulin resistance in a rat model of alcoholic liver disease. Journal of hepatology. 2011;54(1):164–72. pmid:20961644
  44. 44. Speidel D. The role of DNA damage responses in p53 biology. Archives of toxicology. 2015;89(4):501–17. pmid:25618545
  45. 45. Hussain SP, Amstad P, He P, Robles A, Lupold S, Kaneko I, et al. p53-induced up-regulation of MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer research. 2004;64(7):2350–6. pmid:15059885
  46. 46. Mazumder S, Plesca D, Almasan A. Caspase-3 activation is a critical determinant of genotoxic stress-induced apoptosis. Apoptosis and Cancer: Springer; 2008. p. 13–21.
  47. 47. Chen YC, Lin-Shiau SY, Lin JK. Involvement of reactive oxygen species and caspase 3 activation in arsenite-induced apoptosis. Journal of cellular physiology. 1998;177(2):324–33. pmid:9766529
  48. 48. Tyas L, Brophy VA, Pope A, Rivett AJ, Tavarvargy . 1998;177(2):-3 activation during apoptosis revealed using fluorescence-resonance energy transfer. EMBO reports. 2000;1(3):266–70. pmid:11256610
  49. 49. Garrido C, Galluzzi L, Brunet M, Puig P, Didelot C, Kroemer G. Mechanisms of cytochrome c release from mitochondria. Cell Death & Differentiation. 2006;13(9):1423–33. pmid:16676004
  50. 50. Yuan J, Murrell GA, Trickett A, Wang M-X. Involvement of cytochrome c release and caspase-3 activation in the oxidative stress-induced apoptosis in human tendon fibroblasts. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2003;1641(1):35–41.
  51. 51. Dudkina NV, Eubel H, Keegstra W, Boekema EJ, Braun H-P. Structure of a mitochondrial supercomplex formed by respiratory-chain complexes I and III. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(9):3225–9. pmid:15713802