scholarly journals Cell Death and Induced p53 Expression in Oral Cancer, HeLa, and Bone Marrow Mesenchyme Cells under the Exposure to Noncontact Electric Fields

2017 ◽  
Vol 4 (3-4) ◽  
pp. 161-170 ◽  
Author(s):  
Sahudi Abdul Mujib ◽  
Firman Alamsyah ◽  
Warsito Purwo Taruno

Background: p53 acts as a transcription factor to regulate the expression of genes that modulate various cellular activities. The proliferation of cancer cells has been inhibited under the exposure to low-intensity (18 peak-to-peak voltage) and intermediate-frequency (100 KHz) electric fields generated between 2 capacitive electrodes. Therefore, the aims of this study were to observe the molecular mechanism of cell death caused by noncontact electric field exposure and to determine whether p53 protein can serve as a biomarker for this exposure or not. Methods: Oral squamous cell carcinoma, HeLa, and bone marrow mesenchyme cells were exposed to noncontact electric fields of Electro-Capacitive Cancer Therapy (ECCT) for 24 h. To observe the mechanism of cell death caused by ECCT, immunocytochemistry of p53 was performed, and the p53 expression was evaluated using immunoreactive score (IRS) calculation. Results: Electric field exposure by ECCT increased the percentage of dead cells in oral cancer cells (18.39%), HeLa cells (6.60%), and bone marrow mesenchyme cells (34.05%) with statistical significance using the independent t test compared to each control group. The IRS of p53 in oral cancer, HeLa, and bone marrow mesenchyme cultures were 10.50, 11.25, and 4.94, respectively. Conclusion: The high IRS shown in the treated oral cancer and HeLa culture cells may suggest that p53 expression in these culture cells is associated with the cell death mechanism induced by the exposure to noncontact electric fields, and the increased cell death in these culture cells may correlate with the IRS.

2021 ◽  
Vol 11 (4) ◽  
pp. 179-203
Author(s):  
Asaad Shemshadi ◽  
Pourya Khorampour

Facilities and buildings installed nearby high voltage equipment and electric field exposure is always a serious threat to the health of organisms and can have a significant impact on the functioning of sensitive and vital organs such as the heart and brain. Therefore, it is necessary to study the electromagnetic field value in these areas to control the intensity and restrict the induced value regarding to international recommendations. In this paper, the effects of 230KV transmission line electric fields on the environment are examined by proper FEM software.The model under consideration in this project is a four story building adjacent to the 230KV transmission line.At first, the distance between the building and high voltage transmission lines and its relationship to the intensity of the electric field is examined, and then the intensity of the electric field is compared to the standards of the International Commission on Non Ionizing Radiation Protection (ICNIRP). To continue, in places where the electric field exceeds the standard level value, solutions to reduce the intensity of the electric field to the tolerable value have been proposed.The first solution is to use a metal shield around the building as a Faraday cage, which weakens the potential for electric field value by creating an enclosed surface, the reduction rate is 4700%,both complete cage shape and incomplete cage shapes are considered in this study which reduces the exposure value to 62.5% of its initial value. The second approach to reducing the electric field is to use protective conductor paints against electromagnetic fields. In the following study, the effect of using trees as a barrier against electromagnetic radiation will be examined. Finally, the three proposed solutions are compared in terms of environmental constraints, economic justification, and the reduction in electric field value.


2020 ◽  
Author(s):  
L. Martinez ◽  
A. Dhruv ◽  
L. Lin ◽  
E. Balaras ◽  
M. Keidar

AbstractThis paper presents a numerical model to investigate the deformation of biological cells by applying external electric fields operating at or near cell resonant frequencies. Cells are represented as pseudo solids with high viscosity suspended in liquid media. The electric field source is an atmospheric plasma jet developed inhouse, for which the emitted energy distribution has been measured.Viscoelastic response is resolved in the entire cell structure by solving a deformation matrix assuming an isotropic material with a prescribed modulus of elasticity. To investigate cell deformation at resonant frequencies, one mode of natural cell oscillation is considered in which the cell membrane is made to radially move about its eigenfrequency. An electromagnetic wave source interacts with the cell and induces oscillation and viscoelastic response. The source carries energy in the form of a distribution function which couples a range of oscillating frequencies with electric field amplitude.Results show that cell response may be increased by the external electric field operating at or near resonance. In the elastic regime, response increases until a steady threshold value, and the structure moves as a damped oscillator. Generally, this response is a function of both frequency and magnitude of the source, with a maximum effect found at resonance. To understand the full effect of the source energy spectrum, the system is solved by considering five frequency-amplitude couplings. Results show that the total solution is a nonlinear combination of the individual solutions. Additionally, sources with different signal phases are simulated to determine the effect of initial conditions on the evolution of the system, and the result suggests that there may be multiple solutions within the same order of magnitude for elastic response and velocity. Cell rupture from electric stress may occur during application given a high energy source.SignificanceCold atmospheric plasma jets (CAPJs) have been widely researched for their potential applications in cancer therapy. Existing research has focused mainly on the ability of CAPJs to deliver a mixture of reactive species which can be absorbed by cancer cells and induce cell death. The objective of our study is to investigate the mechanical effect of CAPJ electromagnetic (EM) waves on interacting cells. By coupling the EM waves associated with plasma frequency and cell viscoelastic response, we have developed a numerical tool to investigate cell damage either by mechanical or thermal loads. This work is motivated by the promise of EM waves to function as a sensitizing agent for cancer cells in preparation for chemotherapy.


Author(s):  
Rosa Huang Liu ◽  
Shih-Pin Chen ◽  
Tsong-Ming Lu ◽  
Wei-Yu Tsai ◽  
Chung-Hung Tsai ◽  
...  

Author(s):  
Hadi Shafiee ◽  
Rafael V. Davalos

Irreversible electroporation (IRE) is a method to kill cells by exposing the cell to intense electric field pulses[1]. It is postulated that the lipid bilayer rearranges to create permanent defects in the cell membrane which eventually leads to cell death via necrosis[1]. We postulate that the recurrence of cancer for patients treated for the disease would be minimized if their blood was monitored using a microdevice which would destroy existing or new exfoliated cancer cells. Dielectrophoresis (DEP) is the motion of polarizable particles that are suspended in an electrolyte when subjected to a spatially nonuniform electric field [2]. Insulator-based DEP uses insulating structures rather than electrode arrays to produce the nonuniform fields needed to drive DEP. We hypothesize that iDEP can enable the selective IRE of a particular cell type within a microfluidic platform. This manuscript demonstrates through modeling the feasibility of coupling iDEP with IRE using an AC field with a DC offset. Such a platform could be used to selectively destroy isolate cancer cells while not affecting normal cells.


2014 ◽  
Vol 15 (16) ◽  
pp. 6939-6944 ◽  
Author(s):  
Hye-Yeon Han ◽  
Hyungwoo Kim ◽  
Sung-Hee Jeong ◽  
Do-Seon Lim ◽  
Mi Heon Ryu

2020 ◽  
Author(s):  
Qingyuan Feng ◽  
Xuzhao Bian ◽  
Xuan Liu ◽  
Ying Wang ◽  
Huiting Zhou ◽  
...  

Abstract Background: Based on its low toxicity, arginine starvation therapy has the potential to cure malignant tumors that cannot be treated surgically. The Arginine deiminase (ADI) gene has been identified to be an ideal cancer-suppressor gene. ADI expressed in the cytosol displays higher oncolytic efficiency than ADI-PEG20 (Pegylated Arginine Deiminase by PEG 20,000). However, it is still unknown whether cytosolic ADI has the same mechanism of action as ADI-PEG20 or other underlying cellular mechanisms. Methods: The interactions of ADI with other protein factors were screened by yeast hybrids, and verified by co-immunoprecipitation and immunofluorescent staining. The effect of ADI inhibiting the ferritin light-chain domain (FTL) in mitochondrial damage was evaluated by site-directed mutation and flow cytometry. Control of the mitochondrial apoptosis pathway was analyzed by Western Blotting and real-time PCR experiments. The effect of p53 expression on cancer cells death was assessed by siTP53 transfection. Chromatin autophagy was explored by immunofluorescent staining and Western Blotting. Results: ADI expressed in the cytosol inhibited the activity of cytosolic ferritin by interacting with FTL. The inactive mutant of ADI still induced apoptosis in certain cell lines of ASS- through mitochondrial damage. Arginine starvation also generated an increase in the expression of p53 and p53AIP1, which aggravated the cellular mitochondrial damage. Chromatin autophagy appeared at a later stage of arginine starvation. DNA damage occurred along with the entire arginine starvation process. Histone 3 (H3) was found in autophagosomes, which implies that cancer cells attempted to utilize the arginine present in histones to survive during arginine starvation. Conclusions: Mitochondrial damage is the major mechanism of cell death induced by cytosolic ADI. The process of chromatophagy does not only stimulate cancer cells to utilize histone arginine but also speeds up cancer cell death at a later stage of arginine starvation.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 756-756
Author(s):  
Jayakumar R Nair ◽  
Louise M Carlson ◽  
Kelvin P Lee

Abstract Despite novel chemotherapeutic regimens, myeloma patients invariably develop drug resistance and eventually die due to disease relapse, highlighting the necessity for newer therapies to extend survival. Interactions with bone marrow stroma are essential for drug resistance and survival in myeloma (MM). Not much is known about the specific cell types or molecular interactions that mediate this. Clinical studies have shown that expression of CD28 (the prototypic T cell costimulatory molecule) on MM correlates with disease progression. Our recent publication and abstracts (ASH2012 #722, ASH2011 #147, ASH2010 #132) show that interaction with dendritic cells (DC) in the bone marrow provide pro-survival signals to MM through CD28 via interactions with its ligands CD80/CD86 on DC or CD86 on other MM. Clinical studies have separately shown that CD28 or CD86 expression on MM portends poor survival in myeloma patients. In our previous ASH abstracts, we have shown that blocking MM CD28 interactions with CD80/CD86 on DC or other MM using novel reagents such as CTLA4Ig (Abatacept®, a recombinant fusion protein between CTLA4 and human IgFc) or blocking αCD28(Fab) fragments can reduce CD28 mediated survival in MM. In contrast, activating CD28 on MM with agonistic antibodies improves survival against different therapeutics or serum starvation. Another mechanism by which cancer cells survive exogenous stress is via redox regulation and our data that show that BSA (a known scavenger of reactive oxygen species (ROS)) can alleviate CTLA4Ig mediated sensitization of MM cells (Fig 1) suggest involvement of redox regulation in CD28 mediated MM survival. Intrinsic oxidative stress is a hallmark of cancer and is associated with abnormal cancer growth and progression. Others have reported that cancer cells adapt to intrinsic oxidative stress by developing enhanced anti-oxidant capacity, and are more resistant to exogenous stress. Literature also suggests that cancer cells that have higher intrinsic oxidative stress are also more likely to be sensitive to any disruption of redox regulation than normal cells. In MM, flow cytometric analysis using ROS dye CM-DCFDA show significantly higher (8-9 fold) basal ROS levels in the drug resistant U266 as opposed to the drug sensitive cell line MM.1S (Fig 2). Thioredoxin (TRX1) is a key ROS induced anti-oxidant protein essential for redox regulation and survival in many types of cancers. But not much is known about its role in myeloma survival. Interestingly, gene expression analysis of public datasets of plasma cells from normal, MGUS and myeloma patients show significant increases in the levels of TRX1 with disease progression. Further, within the myeloma patient group, TRX1 levels were significantly higher in the “relapsed” group relative to new or smoldering MM groups (Fig 3). In contrast, the levels of thioredoxin-interacting protein (TXNIP), a negative regulator of thioredoxin was significantly lower in myeloma patients and in the relapsed group, compared to normal or new diagnosed patients respectively (Fig 3). Expression trends for TRX1 and TXNIP were inversely correlated across patient groups. While CD28 activation in MM can alleviate drug induced cell death in myeloma, it could not overcome cell death induced by the specific TRX1 inhibitor PX-12 (Biomira Inc, currently under Phase 2 clinical trials for pancreatic cancer). Moreover, viability assays show that high-ROS cell line U266 was 4 fold more sensitive to PX-12 than the low-ROS MM.1S cells which is interesting since U266 is drug resistant and is thus representative of relapsed myeloma. ROS assays with PX-12 on MM show rapid dose dependent increases in ROS levels in U266 cells, while it was much lower in MM.1S suggesting a rationale for the higher sensitivity of U266. This was reversed when a ROS scavenger N-acetyl cysteine (NAC) was added. NAC also completely abrogated PX-12 mediated apoptosis in both myeloma cell lines U266 and MM.1S suggesting that PX-12's activity was solely via disruption of redox regulation. Our data reveal an important redox regulatory mechanism mediated by thioredoxin which play a supportive role for CD28 mediated survival in myeloma and disease relapse, disruption of which could selectively target relapsed drug resistant myeloma. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi120-vi120 ◽  
Author(s):  
Dongjiang Chen ◽  
Nagheme Thomas ◽  
Jie Ren ◽  
Son Le ◽  
Mathew Sebstian ◽  
...  

Abstract OBJECTIVES Glioblastoma (GBM) is the most common and deadliest malignant brain cancer in adults. Tumor Treating Fields (TTFields) was approved in combination with adjuvant temozolomide chemotherapy for newly diagnosed GBM patients. The addition of TTFields resulted in a significant improvement in overall survival. TTFields are low-intensity alternating electric fields that are thought to disturb mitotic macromolecules’ assembly. In many patients, a transient stage of increased peritumoral edema is often observed early during TTFields treatment followed subsequently by objective radiographic responses, suggesting that a major component of therapeutic efficacy by TTFields may be an immune mediated process. We hypothesize that TTFields activate the immune system by triggering pyroptosis and type I Interferon (IFN) response. METHODS A panel of GBM cell lines were treated with TTFields at the clinically approved frequency of 200 kHz using an in vitro TTFields system. Cells were analyzed for the production of micronuclei and activation of both pyroptosis and STING pathways using immunostaining, quantitative PCR, ELISA and cytometry. RESULTS TTFields resulted in a significantly higher rate of micronuclei structures released into the cytoplasm, which were co-localized with two upstream dsDNA sensors AIM2 and cGAS. TTFields-activated micronuclei-dsDNA sensor complexes led to i) induction of pyroptotic cell death, as measured by LDH release assay, and through AIM2-recruited caspase1 and cleavage of pyroptosis-specific Gasdermin D; and ii) activation of STING pathway leading to the release of type I IFNs and pro-inflammatory cytokines downstream of the NFκB pathway. In a co-culture experiment of bone marrow cells with cells/supernatants obtained from GBM cells treated with TTFields, GBM cells depleted of AIM2 and STING failed to induce bone marrow cells. CONCLUSIONS These results provide compelling evidence that TTFields activates the innate immune system in GBM cells, and a strong rationale for combining TTFields with immune checkpoint inhibitors to create a therapeutic synergy.


Sign in / Sign up

Export Citation Format

Share Document