Indian Journal of Animal Research

  • Chief EditorK.M.L. Pathak

  • Print ISSN 0367-6722

  • Online ISSN 0976-0555

  • NAAS Rating 6.50

  • SJR 0.263

  • Impact Factor 0.4 (2024)

Frequency :
Monthly (January, February, March, April, May, June, July, August, September, October, November and December)
Indexing Services :
Science Citation Index Expanded, BIOSIS Preview, ISI Citation Index, Biological Abstracts, Scopus, AGRICOLA, Google Scholar, CrossRef, CAB Abstracting Journals, Chemical Abstracts, Indian Science Abstracts, EBSCO Indexing Services, Index Copernicus
Indian Journal of Animal Research, volume 55 issue 3 (march 2021) : 340-346

In vitro Cytotoxicity, Apoptosis, Effects on Cell Cycle Kinetics and Schedule-Dependent Effects Induced by Paclitaxel on C6 and CHO-K1 Cell Lines

Mohammad Aamir Bhat1,*, Chandresh Varshneya1, Pallavi Bhardwaj1, Rajendra Damu Patel1, Ashok Kumar Panda1
1DGCN College of Veterinary and Animal Sciences, CSKHPKV, Palampur-176 062, Himachal Pradesh, India.
Cite article:- Bhat Aamir Mohammad, Varshneya Chandresh, Bhardwaj Pallavi, Patel Damu Rajendra, Panda Kumar Ashok (2020). In vitro Cytotoxicity, Apoptosis, Effects on Cell Cycle Kinetics and Schedule-Dependent Effects Induced by Paclitaxel on C6 and CHO-K1 Cell Lines . Indian Journal of Animal Research. 55(3): 340-346. doi: 10.18805/ijar.B-3955.
Exposure of C6 and CHO-K1 cells to different concentrations of the antineoplastic drug paclitaxel resulted in a loss of cellular viability. The percentage of surviving cells fell significantly after 48 hours of treatment and IC50 values observed were between 0.5 to 0.75 and 0.25 to 0.75 µg/ml in C6 and CHO-K1 cells, respectively. No significant cytotoxicity was observed after 24 hours of treatment and cells incubated at higher concentrations of paclitaxel showed increased survivability. Paclitaxel induced apoptosis by caspase 3/7 activation and caused accumulation of cells in the G2/M phase of the cell cycle. Upon fluorescent microscopy, both the cell lines lost the morphology, confluence and adherence at 24 hours but effects were much more pronounced at 48 hours of treatment. The in vitro data suggested that paclitaxel is highly effective when there is prolonged exposure of tumor to the drug rather than increasing the intratumoral or biophasic concentration of the drug. 
Paclitaxel is a miraculous drug, one of most extensively studied, complex semi-synthetic diterpine alkaloid, an antineoplastic agent obtained primarily from the European Yew (Taxus Baccata) and the Pacific Yew (Taxus Brevifolia), widely used against lung, advanced breast and ovarian carcinomas (Bhat et al., 2016). It is also being employed for the treatment of squamous cell carcinomas of head and neck, prostrate, endometrial, gastroesophageal cancers and recent studies have shown that paclitaxel can also be used for the treatment of leukemia, lymphoma and sarcoma Khanna et al., (2015).
 
Paclitaxel is a microtubule-stabilizing anticancer agent, inducing mitotic arrest by suppression of microtubule dynamics (Schiff et al., 1979). The over stabilization of microtubules results in the inhibition of normal re-organization of the network, leading to the cytotoxic effects in a subset of the arrested population (Diaz et al., 1993). However, recent studies have shown that paclitaxel does not acquire sufficient intra-tumoral concentrations to cause mitotic arrest and it has been observed that the cytotoxic effects may be due to chromosome misaggregation on multipolar spindles (Beth, 2014). 
 
Taxanes have revolutionized the cancer chemotherapy and have shown great results when used in combination with other cytotoxic agents but the cytotoxic potential of paclitaxel against gliomas has not been elucidated conclusively (Bhat et al., 2016). Gliomas account for majority of primary malignant brain tumors and are associated with very poor survival rates. It is of utmost importance to formulate formulations achieving profound cytotoxic concentrations inside glioma tumoral cells, with minimal damage to normal cells (Bhat et al., 2016).
 
The present study was undertaken to study in vitro cytotoxic effects, apoptosis, effects on cell cycle kinetics and schedule dependent effects induced by paclitaxel on C6 (Rattus norvegicus glioma cell line) and CHO-K1 cell lines (Chinese Hamster ovarian carcinoma).
Paclitaxel
 
Paclitaxel (10 mg, Catalogue No. RM9750) was obtained from HIMEDIA and stored at 2-8°C.
 
Cell lines and Cell culture
 
C6 (glioma cells) cells were obtained from National Centre for Cell Science, Pune-India and cultured in DMEM (Sigma Aldrich) supplemented with 10% heat inactivated fetal bovine serum and 1% antibiotic-antimycotic (Sigma Aldrich). CHO- K1 (Chinese hamster ovary cells) cells were obtained from CSIR-Indian Institute of Integrative Medicine, Jammu-India and were cultured in HAM’S-F12 medium (Sigma Aldrich), supplemented with 10% heat inactivated fetal bovine serum (Sigma Aldrich) and 1% antibiotic-antimycotic (Sigma Aldrich). Both the cell lines were maintained at 37°C in 5% CO2 humidified atmosphere (Katoch et al., 2012).
 
Sulforhodamine B assay
 
The level of cytotoxicity was measured using the Sulforhodamine B (SRB) method (Walia et al., 2012). The viable cells were seeded in the growth medium (100 μL) into 96 well microtiter plates (5X104 cells per well) and allowed to adhere overnight. The test sample, paclitaxel, was dissolved in DMSO and several dilutions (0.25, 0.50, 0.75, 1.25, 2.50 and 5 μg/ml) of the test compound in 100 μl of respective complete media was added to the wells. The cell culture alone supplemented with complete medium was used as a negative control, while as vinblastine (1μM) was used as a positive control for the assay. After 24 and 48 hours, 50 μl of 50% trichloroacetic acid was added to the wells and the plates were incubated at 4°C for one hour. The TCA treated cells were washed extensively with water and air dried. Subsequently, 100 μL of the SRB solution (in 1% Acetic acid) was added to each well at room temperature. After standing for 30 min, the wells were washed 5-6 times with 1% Acetic Acid and air dried. The bound dye was dissolved in 10 mM tris base (100 μl/well). The absorbance was measured using microplate reader (BioTeK Synergy H1 Hybrid Reader) at the wavelength of 540 nm (Joshi et al., 2012). The growth inhibition rate was calculated using the following formula:
 
           % Cell inhibition = 100 - {(At-Ab)/(Ac-Ab)} x 100
 
 
Where, At, Ab and Ac represent absorbance value of test compound, blank and control, respectively.
 
Apoptotic (Caspase 3/7 activity) assay
 
The cells were plated in 96-well plates at 2×104 density. After 3 hours of treatment of cells, caspase activity was detected against C6 and CHO-K1 cell lines using different concentrations of paclitaxel (0.50, 0.75, 1.25 μg/ml against CHO-K1and 1.0, 1.5 and 2.5 μg/ml against C6). The total caspase activity was detected using the Caspase-Glo® 3/7 Assay kit (Promega, Catalogue No. G8090). The medium alone was used as a negative control for the activity, whereas the cell cultures exposed only to DMSO (1%) was used as a positive control. The activity was measured according to the supplier’s instructions provided with the kit. Caspase-3/7 activity was measured in net relative luminescence units (RLU) of respective incubated cells in the 96-well plates and was measured using microplate reader (BioTeK Synergy H1 Hybrid Reader) as per the recommended protocol. Following formula was used to calculate caspase 3/7activity in relative luminescence units (RLU):
 
 RLU = Luminescence (samples) – Luminescence (blank)

Cell cycle analysis
 
Cell cycle analysis was done by Muse™ Cell Cycle Kit, according to the manufacturer’s instructions (Catalogue No. MCH100106). Ethanol-fixed cells were treated with a premixed Muse™ Cell Cycle Reagent and incubated at room temperature for 30 minutes. The concentrations of paclitaxel applied in C6 and CHO-K1 cells were 0.75 and 0.5 μg, respectively.
 
Morphological changes
 
The morphological changes of C6 and CHO-K1 cells treated with paclitaxel for 24 and 48 hours were observed by using inverted fluorescent microscope (Nikon Eclipse Ti) at 10X magnification.
Sulforhodamine B assay
 
Paclitaxel showed highest inhibition on C6 cells at concentration of 1.25 μg/ml and on CHO-K1 cells at 0.5 μg/ml after 48 hours of treatment. No significant changes were obtained after 24 hours in both the cell lines. The IC50 values of Paclitaxel were observed between 0.5 to 0.75 µg/ml in case of C6 cells and 0.25 to 0.75 µg/ml in CHO-K1 cells. The results are depicted in Table 1 and 2.

Table 1: Cytotoxic effect of paclitaxel on C6 cell line after 24 and 48 hours of treatment.



Table 2: Cytotoxic effect of paclitaxel on CHO-K1 cell line after 24 and 48 hours of treatment.



In both cell lines the percentage of cytotoxicity increased with increase in the time dependent exposure of cells to paclitaxel. This schedule dependent effect of paclitaxel may be because longer durations of drug exposure may allow greater proportion of cells to cycle into the susceptible phase (Georgiadis et al., 1997). The results showed that cell death occurs in cells with slower kinetics rather than using increasing concentrations of paclitaxel suggesting that cells must spend sufficient time in presence of paclitaxel, as longer durations of drug exposure may induce lethality in the greater proportion of cells (Katherine et al., 2013) and previous studies have shown that prolonged exposure of clinically relevant concentrations of paclitaxel may lead to the formation of chromosome misaggregation on multipolar spindles, causing lethality (Lauren et al., 2014). The results of this assay are in agreement with previous studies that analyzed the growth inhibiting effects of paclitaxel and found concentration of paclitaxel needed to inhibit tumour cell growth by 50% (IC50) was typically in the nanomolar/L range (Maria, 2014).
 
Apoptotic (Caspase-3/7 activity) assay
 
Caspases are a family intracellular proteases (cysteinyl-dirested aspartate specific proteases) (Riedl and Shi, 2004) that regulate the process of programmed cell death in response to proapoptotic signals. According to their mechanism of action, caspases can be classified as initiator (caspase -8 and -9) and executioner caspases (caspase -3.-6 and -7). Initiator caspases activate executioner caspases that eventually leads to apoptosis. Activation of caspases -3 and -7 is regarded as a hallmark of apoptosis (Mcllwain et al., 2013 and Portera and Janike, 1999).
 
Paclitaxel showed highest relative luminescence unit at the concentration of 2.5 μg/ml against C6 and at 1.25 μg/ml against CHO-K1 cell lines. The results depicted in Table 3 and 4, show significant activation of caspases and are in agreement with Byrne et al., (2006), who found that paclitaxel elicited a caspase response at the IC50 value of 20-30 µM in normal human mammalian epithelial cells (HMECs) and mammary tumor cell line (MDA-MB-231). In this present study, caspases-3/7 were dramatically activated at 48-72 hours with increasing paclitaxel concentration and enhanced activity of caspases-3/7 was evidently verified by the measurement of the cleavage of poly(ADP-ribose) polymerase (PARP). The significant increase in the activity of caspases-3/7 suggested the schedule dependent induction of apoptosis (Jelinek et al., 2015). However, previous studies have shown that caspase activation may not be the only mechanism of apoptosis induced by paclitaxel and may also be dependent upon the intracellular concentrations of the drug, for instance, apoptosis may occur by p53 up-regulation instead of following the caspase pathway Kim et al., (2013), especially at low concentrations at which caspase reactions are not significantly active in inducing apoptosis (Tan et al., 2002).

@tabole3

Table 4: Caspase activity of paclitaxel against CHO-K1 cancer cell line.


 
Cell cycle analysis
 
Muse™ Cell Cycle Assay utilizes the differential staining of cells based on the DNA content. The nuclear DNA stain, prodidium iodide, intercalates with the DNA causing fluorescence. Cells in Go/G1 state contain diploid set of chromosomes and as the cells begin cycling the fluorescence increases and doubles in the G2/M phase, followed by the cellular division (Ho et al., 2004).
 
Paclitaxel at concentration of 0.75 μg/ml caused an accumulation of the cells in G2/M phase by about 41% and that of control by about 20.1%, in C6 cells. 0.5 μg/ml concentration was applied to CHO-K1 cells and it caused an accumulation of cells in G2/M phase by about 57.7% and that of control by about 25.6%. These results suggest that G2/M arrest and a decrease in the population of cells in G1 phase, was induced by paclitaxel in both the cell lines. These results are in agreement with previous studies, for instance, Juliet et al., (2014) showed 25 nM and 50 nM of paclitaxel induced G2M blocked in MDA-MB-231 and ZR75-1 cells.
 
The concentrations used in the given experimental were sufficient to induce caspase activation and interact significantly with the spindle-proteins leading to mitotic arrest. However, previous studies also reveal that paclitaxel can induce Garrest at low intracytoplasmic concentrations and may be because of Bcl-2 phosphorylation (Kim et al., 2013) and chromosome misaggregation on multipolar spindles (Lauren et al., 2014).
The results are depicted in Table 5 to 8 and Fig 1 and 2.

Table 5: Cell cycle kinetics of untreated C6 cell line.



Table 6: Cell cycle kinetics of treated C6 cell line.



Table 7: Cell cycle kinetics of untreated CHO-K1 cell line.



Table 8: Cell cycle kinetics of treated CHO-K1 cell line.



Fig 1: DNA content index of treated and untreated C6 cell line.



Fig 2: DNA content index of treated and untreated CHO-K1 cell line.


 
Morphological changes
 
The morphological changes in C6 and CHO-K1 cell lines treated with 0.75 and 0.5 μg/ml of paclitaxel, respectively, for 24 and 48 hours, were observed, as determination of key adherent cell culture characteristics such as confluency morphology and cell density are necessary for the evaluation of experimental outcomes.
 
Paclitaxel showed increased potential of cytotoxicity in a time dependent manner on both the cell lines. Untreated C6 cell line represented normal neuronal morphology, confluency and cell density (plate 1) however on treatment for 24 hours cell line lost its morphology and decreased confluency and cell density was observed (plate 2). The effects were much more pronounced at 48 hours (plate 3) of treatment.
CHO-K1 is an epithelial type of cell line (plate 4), so its cell size is smaller than C6, as evident from the respective untreated plates. The treatment for 24 hours caused a decrease in cell line density but retained some ability of adherence with slight reduction in confluency (plate 5). It may be because of the fact that concentration of paclitaxel used was less than that of used in C6. CHO-K1 showed much more pronounced decrease in adherence, confluency and density at 48 hours (plate 6).

Plate 1: UNTREATED C6.



Plate 2: C6_24 hr.



Plate 3: C6_48 hr.



Plate 4: UNTREATED CHO-K1.



Plate 5: CHO-K1_24 hr.



Plate 6: CHO-K1_48 hr.


 
Adherent property is an important feature of C6 and CHO-K1 and upon treatment both the cell lines lost their adherence thereby decreasing the cell density and confluency indicating cytotoxic effects on C6 and CHO-K1 (Jaccard et al., 2013).
Paclitaxel showed highest inhibition on C6 cells at concentration of 1.25 μg/ml and on CHO-K1 cells at 0.5 μg/ml after 48 hours of treatment. No significant changes were obtained after 24 hours in both the cell lines. Cells incubated in high concentrations of paclitaxel had increased survivability compared with cells treated with lower concentrations of the drug. The IC50 values of Paclitaxel were observed between 0.5 to 0.75 µg/ml in case of C6 cells and 0.25 to 0.75 µg/ml in CHO-K1 cells. Apoptosis was screened by employing Caspase-Glo® 3/7 Assay kit. Paclitaxel showed highest RLU at concentration of 2.5 μg/ml against C6 and at 1.25 μg/ml against CHO-K1 cell line, clearly indicating paclitaxel leads apoptosis by caspase 3/7 activation. Cell cycle analysis was done by MuseCell Cycle Kit and G2/M phase arrest was observed in both the cell lines. Upon fluorescent microscopy, both the cell lines lost their morphology, confluency and adherence after 24 hours but the effects were much more pronounced after 48 hours of treatment.
 
The in vitro data suggests that paclitaxel is highly effective when there is prolonged exposure of tumour to the drug rather than increasing the intrumoral or biophasic concentration of drug.
I would like to acknowledge the financial, academic and technical support of DCGN, College of Veterinary and Animal Sciences, CSKHPKV and its staff, particularly in the award of a Postgraduate Research Studentship that provided the necessary support to conduct the research.
The authors do not present any conflict of interest.

  1. Beth, A.W. (2014). How Taxol/Paclitaxel kills Cells. Molecular Biology of the Cell. 25(18): 2677-2681.

  2. Bhat, M.A., Varshneya, C., Bhardwaj, P., Patil, R.J. (2016). Paclitaxel-Development, properties, toxicity/safety evaluation and the scope of improvement. IOSR Journal of Biotechnology and Biochemistry. 2(4):40-48.

  3. Byrne, A.T., Miller, I., Hall, M., Holmes, L., O’Shea, D.F., Wilde, C., Gallagher, W.M. (2006). New cell-based assay for comparison of caspase 3/7 activity in normal versus tumor cell populations. Experimental and Molecular Therapeutics. 10(2):33-39.

  4. Diaz, J.F. and Andreu, J.M. (1993). Assembly of purified GDP-tubulin into microtubules induced by taxol and taxotere: reversibility, ligand stoichiometry and competition. Biochemistry. 32: 2747-2755.

  5. Georgiadis, M.S., Russell, E.K., Gazdar, A.F., Johnson, B.E. (1997). Paclitaxel cytotoxicity against human lung cancer cell lines increases with prolonged exposure durations. Clinical Cancer Research. 3:449-454.

  6. Ho, J., Gillis, K., Fishwild, D. (2004). A simple and robust system for determining cells cycle distribution: the Guava cell cycle assay. Guava Technologies Application Note.

  7. Jaccard, N., Grifûn, L.D., Keser, A., Macown, R.J., Super, A., Veraitch, F.S., Szita, N. (2013). Automated method for the rapid and precise estimation of adherent cell culture characteristics from phase contrast microscopy images. Biotechnology and Bioengineering. 111(3):504-517.

  8. Jelinek, M., Balusikowa, K., Schmiedlova, M., Nemcova-Furstova, V., Sramek, J., et al. (2015). The role of individual caspases in cell death induction by taxanes in breast cancer cells. Cancer Cell International. 15(1):8.

  9. Joshi, R., Sood, S., Dogra, P., Mahendru, M., Kumar, D., Bhangalia, S., Pal, H.C., Kumar, N., et al (2012). In vitro cytotoxicity, antimicrobial and metal-chelating activity of triterpene saponins from tea seed grown in Kangra valley. India Medicinal Chemistry Research. 22:4030-4038.

  10. Juliet, K., Melanie, S., Nicola, L., Karen, J.T., Linda, L., Carrie, A.C., Maryou, L., Alan, M., et al. (2014). Molecular characterisation of isogenic taxane resistant cell lines identify novel drivers of drug resistance. BMC Cancer. 14:762.

  11. Katherine, L., Shundong, C., Yuehua, M., Jen, W.C. (2013). Synergistic effect of paclitaxel and epigenetic agent phenethyl isothiocyanate on growth inhibition, cell cycle arrest and apoptosis in breast cancer cells. Cancer Cell International. 13:10. DOI: 10.1186/1475-2867-13-10.

  12. Katoch, D., Kumar, D., Sharma, U., Kumar, N., Padwad, Y.S., Lal, B., Singh, B. (2012). Zephgrabetaine: A new betaine type Amaryllidaceae alkaloid from Zephyranthes grandiflora. Natural Product Communications. 8(2):160-164.

  13. Khanna, C., Rosenberg, M., Vail, D.M. (2015). A Review of paclitaxel and novel formulations including those suitable for use in dogs. Journal of Veterinary Internal Medicine. 29(4): 1006-1012.

  14. Kim, J.H., Yoon, E.K., Chung, H.J., Park, S.Y., Hong, K.M., Lee, C.H., Lee, Y.S., Choi, K., et al. (2013). P53 acetylation enhances taxol-induced apoptosis in human cancer cells. Apoptosis. 18:110-120.

  15. Lauren, M.Z., Kristen, A.A., Dabin, Y., Gabrielle, B.R., Lee, G.W., Amye, J.T., Ronald, T.R., Mark, E.B., Beth, B.A. (2014). Cytotoxicity of paclitaxel in breast cancer is due to chromosome misaggregation on multipolar spindles. Science Translational Medicine. 26:6(229):229ra43. DOI: 10.1126/scitranslmed.3007965.

  16. Maria Kavallaris. (2014). Cytoskeleton and Human Disease. Human Press. Randwick, Australia.

  17. Mcllwain, D.R., Berger, T., Mak, T.W. (2013). Caspase functions and Cell Death. Cold Spring Harbor Perspectives in Biology. DOI: 10.1101/cshperspect.a008656.

  18. Portera, A.G. and Jänicke, R.U. (1999). Emerging roles of caspase-3 in apoptosis. Cell Death and Differentiation. 6:99-104.

  19. Riedl, S.J. and Shi, Y. (2004). Molecular mechanisms of caspase regulation during apoptosis. Nature Reviews Molecular Cell Biology. 5:897-907.

  20. Schiff, P.B., Fant, J., Horwitz, S.B. (1979). Promotion of microtubule assembly in vitro by taxol. Nature. 277: 665-667.

  21. Tan, G., Heqing, L., Jianbo, C., Ming, J., Yanhong, M., Xianghe, L., Hong, S., Li, G. (2002). Apoptosis induced by low-dose paclitaxel is associated with p53 upregulation in nasopharyngeal carcinoma cells. International Journal of Cancer. 97:168-172.

  22. Walia, M., Mann, T., Kumar, D., Agnihotri, V.K., Singh, B. (2012). Chemical composition and in vitro cytotoxic activity of essential oil of leaves of Malus domestica growing in Western Himalaya (India). Evidence-Based Complementary and Alternative Medicine. 2012: 649727. DOI:10.1155/2012/ 649727.

Editorial Board

View all (0)