scholarly journals Radiation-induced cell cycle perturbations: a computational tool validated with flow-cytometry data

2020 ◽  
Author(s):  
Leonardo Lonati ◽  
Sofia Barbieri ◽  
Isabella Guardamagna ◽  
Andrea Ottolenghi ◽  
Giorgio Baiocco

AbstractCell cycle progression can be studied with computational models that allow to describe and predict its perturbation by agents as ionizing radiation or drugs. Such models can then be integrated in tools for pre-clinical/clinical use, e.g. to optimize kinetically-based administration protocols of radiation therapy and chemotherapy.We present a deterministic compartmental model, specifically reproducing how cells that survive radiation exposure are distributed in the cell cycle as a function of dose and time after exposure. Model compartments represent the four cell-cycle phases, as a fuction of DNA content and time. A system of differential equations, whose parameters represent transition rates, division rate and DNA synthesis rate, describes the temporal evolution. Initial model inputs are data from unexposed cells in exponential growth. Perturbation is implemented as an alteration of model parameters that allows to best reproduce cell-cycle profiles post-irradiation. The model is validated with dedicated in vitro measurements on human lung fibroblasts (IMR90). Cells were irradiated with 2 and 5 Gy with a Varian 6 MV Clinac at IRCCS Maugeri. Flow cytometry analysis was performed at the RadBioPhys Laboratory (University of Pavia), obtaining cell percentages in each of the four phases in all studied conditions up to 72 hours post-irradiation.Cells show early G2-phase block (increasing in duration as dose increases) and later G1-phase accumulation. For each condition, we identified the best sets of model parameters that lead to a good agreement between model and experimental data, varying transition rates from G1- to S- and from G2- to M-phase.This work offers a proof-of-concept validation of the new computational tool, opening to its future development and, in perspective, to its integration in a wider framework for clinical use.Author summaryWe implemented a computational model able to describe how the progression in the cell cycle is perturbed when cells are exposed to ionizing radiation. It is known that radiation causes delays or arrest in cell cycle progression, and also that cells that are in different phases of the cycle at the time of exposure show different sensitivity to radiation. Chemotherapeutic drugs also affect cell cycle, and their action can be phase-specific. These findings can be exploited to find the optimal protocol of a combined radiotherapy/chemotherapy cancer treatment: to this aim, we need to know not only the effectiveness of an agent (dose/drug) in terms of cell killing, but also how surviving cells are distributed in the cell cycle. With the model we present, this information can be reproduced as a function of dose and time after radiation exposure. To test the model performance we measured distributions of cells in different phases of the cycle (using flow-cytometry) for human healthy fibroblast cells exposed to X-rays. The results of this work constitute a first step for further development of our model and its future integration in a tool for pre-clinical/clinical use.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Leonardo Lonati ◽  
Sofia Barbieri ◽  
Isabella Guardamagna ◽  
Andrea Ottolenghi ◽  
Giorgio Baiocco

AbstractCell cycle progression can be studied with computational models that allow to describe and predict its perturbation by agents as ionizing radiation or drugs. Such models can then be integrated in tools for pre-clinical/clinical use, e.g. to optimize kinetically-based administration protocols of radiation therapy and chemotherapy. We present a deterministic compartmental model, specifically reproducing how cells that survive radiation exposure are distributed in the cell cycle as a function of dose and time after exposure. Model compartments represent the four cell-cycle phases, as a function of DNA content and time. A system of differential equations, whose parameters represent transition rates, division rate and DNA synthesis rate, describes the temporal evolution. Initial model inputs are data from unexposed cells in exponential growth. Perturbation is implemented as an alteration of model parameters that allows to best reproduce cell-cycle profiles post-irradiation. The model is validated with dedicated in vitro measurements on human lung fibroblasts (IMR90). Cells were irradiated with 2 and 5 Gy with a Varian 6 MV Clinac at IRCCS Maugeri. Flow cytometry analysis was performed at the RadBioPhys Laboratory (University of Pavia), obtaining cell percentages in each of the four phases in all studied conditions up to 72 h post-irradiation. Cells show early $${\text{G}}_{2}$$ G 2 -phase block (increasing in duration as dose increases) and later $${\text{G}}_{1}$$ G 1 -phase accumulation. For each condition, we identified the best sets of model parameters that lead to a good agreement between model and experimental data, varying transition rates from $${\text{G}}_{1}$$ G 1 - to S- and from $${\text{G}}_{2}$$ G 2 - to M-phase. This work offers a proof-of-concept validation of the new computational tool, opening to its future development and, in perspective, to its integration in a wider framework for clinical use.


1983 ◽  
Vol 38 (3-4) ◽  
pp. 313-318 ◽  
Author(s):  
Rainer Merz ◽  
Friedhelm Schneider

Utilizing centrifugal elutriation, early and late S-phase cells were separated from 4, 8 and 12 h anaerobically cultured Ehrlich Ascites tumor cells strain Karzel. The cytokinetic properties of these fractions after reaeration were studied by flow cytometry and the BrdU-H 33258-technique of flow cytometry. After a 4 h period of anaerobiosis, growth of early S-phase cells is not changed, 8 h deprivation of oxygen causes a delay of cell cycle progression, while the main fraction of 12 h anaerobically treated early S-populations did not divide after reaeration within 24 h. In comparison to early S-phase cells the cell cycle progression of the main fraction of late S-period is accelerated after a 4 h exclusion of oxygen. A fraction of 8 h anaerobically pretreated late S-cells continues to cycle, but a considerable number reinitiates DNA synthesis without preceeding division. Cells with DNA content up to 8 c are detected by flow cytometry. 12 h anaerobically cultured late S-cells do not divide after reaeration, a large number of these cells starts again to synthesize DNA. A considerable part of tetraploid cells retain viability, divide and enter a new cell cycle, another part of the cells disintegrates


Author(s):  
Afza Ahmad ◽  
Irfan A. Ansari

Background:: The carcinogenesis of uterine cervix is predominantly initiated with the consistent infection of human papilloma virus (HPV). Owing to the adverse side effects of standard chemotherapeutics in the treatment of recurrent and metastatic cervical cancer, there is a need for better and effective treatment modality. In this lieu of concern, natural compounds have proven their worthwhile potential against treatment of various carcinomas. Carvacrol is a phenolic monoterpenoid and several reports have suggested its different biological properties including antioxidant, anti-inflammatory and anticancer activity. Objective:: The objective of our present study was to investigate the effect of carvacrol on HPV18+ HeLa cervical cancer cells. Methods:: HeLa cervical cancer cells were cultured and subsequently treated with various doses of carvacrol. Cell viability was assessed via MTT assay. DAPI and Hoechst3342 staining was used to qualitatively analyzed the induced apoptosis. Reactive Oxygen Species (ROS) was estimated by DCFDA staining protocol and quantitatively estimated by flow cytometry. The cell cycle distribution and apoptosis (FITC-Annexin V assay) were analyzed by flow cytometry. Results:: The results of the present study have established that carvacrol strongly suppresses proliferation of cervical cancer cells via caspase-dependent apoptosis and abrogation of cell cycle progression. Furthermore, our preliminary study also demonstrated that carvacrol exhibits synergistic effect with chemotherapeutic drugs (5-FU and carboplatin). These initial findings implicated that natural compounds could reduce the toxic effects of chemotherapeutic drugs. Conclusion:: Therefore, this investigation affirms the anti-cancer potential of carvacrol against cervical cancer cells which could be an appendage in the prevention and treatment of cervical cancer.


1996 ◽  
Vol 16 (4) ◽  
pp. 1500-1508 ◽  
Author(s):  
P C Goswami ◽  
J L Roti Roti ◽  
C R Hunt

Topoisomerase II is a multifunctional protein required during DNA replication, chromosome disjunction at mitosis, and other DNA-related activities by virtue of its ability to alter DNA supercoiling. The enzyme is encoded by two similar but nonidentical genes: the topoisomerase IIalpha and IIbeta genes. In HeLa cells synchronized by mitotic shake-off, topoisomeraseII alpha mRNA levels were found to vary as a function of cell cycle position, being 15-fold higher in late S phase (14 to 18 h postmitosis) than during G1 phase. Also detected was a corresponding increase in topoisomerase IIalpha protein synthesis at 14 to 18 h postmitosis which resulted in significantly higher accumulation of the protein during S and G2 phases. Topoisomerase IIalpha expression was not dependent on DNA synthesis during S phase, which could be inhibited without effect on the timing or level of mRNA expression. Mechanistically, topoisomerase IIalpha expression appears to be coupled to cell cycle position mainly through associated changes in mRNA stability. When cells are in S phase and mRNA levels are maximal, the half-life of topoisomerase IIalpha mRNA was determined to be approximately 30 min. A similar decrease in mRNA stability was also induced by two external factors known to delay cell cycle progression. Treatment of S-phase cells, at the time of maximum topoisomerase IIalpha mRNA stability, with either ionizing radiation (5 Gy) or heat shock (45 degrees C for 15 min) caused the accumulated topoisomerase IIalpha mRNA to decay. This finding suggests a potential relationship between stress-induced decreases in topoisomerase IIalpha expression and cell cycle progression delays in late S/G2.


1995 ◽  
Vol 34 (2) ◽  
pp. 79-83 ◽  
Author(s):  
Eric J. Bernhard ◽  
Amit Maity ◽  
Ruth J. Muschel ◽  
W. Gillies McKenna

Sign in / Sign up

Export Citation Format

Share Document