2018
DOI: 10.3892/ijo.2018.4439
|View full text |Cite
|
Sign up to set email alerts
|

Microwave hyperthermia promotes caspase‑3-dependent apoptosis and induces G2/M checkpoint arrest via the ATM pathway in non‑small cell lung cancer cells

Abstract: Post-operative microwave (MW) hyperthermia has been applied as an important adjuvant therapy to enhance the efficacy of traditional cancer treatment. A better understanding of the molecular mechanisms of MW hyperthermia may provide guided and further information on clinical hyperthermia treatment. In this study, we examined the effects of MW hyperthermia on non-small cell lung carcinoma (NSCLC) cells in vitro, as well as the underlying mechanisms. In order to mimic clinical treatment, we developed special MW h… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

3
17
0

Year Published

2020
2020
2024
2024

Publication Types

Select...
7
1

Relationship

0
8

Authors

Journals

citations
Cited by 23 publications
(20 citation statements)
references
References 44 publications
3
17
0
Order By: Relevance
“…Although the very notion of temperature at this scale and far from a thermodynamical equilibrium is not clear, the measurement of the temperature variations inside the cell during its life cycle could be correlated with the onset of mitosis and with the activity of the mitochondrial network (31,32). Our proposition is also coherent with the fact that hyperthermia can block the cell cycle at the G2/M checkpoint (33).…”
Section: Resultssupporting
confidence: 73%
See 1 more Smart Citation
“…Although the very notion of temperature at this scale and far from a thermodynamical equilibrium is not clear, the measurement of the temperature variations inside the cell during its life cycle could be correlated with the onset of mitosis and with the activity of the mitochondrial network (31,32). Our proposition is also coherent with the fact that hyperthermia can block the cell cycle at the G2/M checkpoint (33).…”
Section: Resultssupporting
confidence: 73%
“…Let us add that the thermal gradient still plays an important role in the division of modern cells because hyperthermia can block the cell cycle just before mitosis, as observed experi-mentally on lung cancer cells (33). This is why we consider our proposition as a first step toward a more detailed model that would take into account other thermodynamical forces, such as the membrane electric potential or ionic concentration differences (34), to explain the fission of protocells.…”
Section: Resultsmentioning
confidence: 89%
“…High-temperature stimulation leads to several physiological responses, including changes in the membrane permeability or cytoskeleton system, alteration in macromolecule synthesis or intracellular signaling, and inhibition of DNA repair [ 10 ]. In lung cancer, hyperthermia treatment induces cell death via cytoskeletal alteration [ 11 ], an increase in caspase-3-dependent apoptosis [ 12 ], or the induction of reactive oxygen species-related autophagy [ 13 ]. In addition, the effects of gemicitabine [ 14 ], paclitaxel/carboplatin [ 15 ], doxorubicin [ 16 ], cisplatin [ 17 ], or even radiotherapy [ 18 ] can be increased when combined with hyperthermia.…”
Section: Introductionmentioning
confidence: 99%
“…As part of a combined treatment with chemo- or radiotherapy, hyperthermia is already approved for routine clinical use in several countries. The radiosensitizing effect is thought to occur due to the improvement of oxygen delivery to the tissue [ 290 , 291 ] and due to interference with the DNA damage response [ 292 298 ]. Cytotoxic effects can be direct and indirect: heat can directly induce necrosis, apoptosis and mitotic catastrophe [ 299 301 ] and with an applied temperature above 42 °C, heat will indirectly kill by inducing vascular damage [ 302 ].…”
Section: Radiotherapy and Tumor Hypoxiamentioning
confidence: 99%