Abstract
Natural products based on their significant anti-cancer potencies have been used in cancer treatment. A natural blend of triterpenoid saponins derived from the horse chestnut (Aesculus hippocastanum L.), has been investigated in various diseases based on its main active ingredient escin. Herein, we examined the potential antiproliferative, proapoptotic, and cytotoxic activities of escin on human skin melanoma (CHL-1) cells. Cytotoxicity of escin was determined by MTT assay. Morphological changes were detected by confocal microscopy and ultrastructural changes by transmission electron microscopy studies. Phosphatidylserine translocation assay, Bcl-2 activation assessment, and oxidative stress analysis were used to determine the cell death mode of the cells. The results showed that escin reduced cell viability in a dose-dependent manner within 24 h of exposure and was highly cytotoxic at lower concentrations (IC50 value 6 μg/mL). Escin inactivated Bcl-2 signaling and triggered apoptosis by increasing the reactive oxygen species and by causing morphological and ultrastructural changes that implicate to the proapoptotic activity. Escin has been found to exert high potential for an anti-cancer drug following further in vitro and in vivo investigations.
Keywords: escin, cancer treatment, melanoma, cytotoxicity, apoptosis
Introduction
Cancer has been defined as a disease derived from uncontrollable and abnormal cell division in different parts of the organism. After the occurrence, it invades surrounding tissues and metastases to other parts through the blood and lymphatic system.1 Cancer is one of the most widespread diseases in the world, with an estimated 14 million new diagnoses and 8.2 million deaths in 2012.2 Many treatment methods, such as surgery, chemotherapy, and radiotherapy, have been used for treating various cancers; yet, the disease remains untreated due to limitations in its treatment methods and side effects of the present medicines.3
Melanoma is the most malignant skin cancer, and it is the sixth and seventh most common cancer in men and women in the United States, respectively. Early-diagnosed melanoma can be cured surgically but metastatic melanoma remains one of the aggressive cancers with high levels of drug resistance.4 Therefore, there is a real need to find effective treatments and agents with slight or no side effects. In the twentieth century, plant-derived products began to be used in cancer treatment.
The first natural product was actinomycin which was isolated in 1964 and used for cancer therapy.5 Other natural products utilized to cure cancer included vancomycin6 and taxol.7 Since then, natural compounds have made up the majority of anti-cancer agents, accounting for 60%.8 Natural products that have the ability to augment chemotherapy efficiency have been used for many years to treat various cancers. Escin is one of the compounds obtained from the seeds of Aesculus hippocastanum (the horse chestnut).9 Horse chestnut is reported to be native to the Balkans but it is also grown in many countries in the World.10 The chestnut tree’s seeds and bark have been used for medical purposes such as treating rheumatism, fevers, bladder and gastrointestinal disorders, leg cramps, and hemorrhoids. Especially seed extracts has been found to have different therapeutic efficacies due to its ingredients such as escin, quercetin kaempferol, and proanthocyanidin.11 Escin is a penta cyclic triterpene that is a saponin blend and can be found in two forms, α and β. The β Escin contains anti-inflammatory and antiedema characteristics, and has been shown to promote apoptotic cell death in various cancer cells.9,12 Escin has been highly investigated in many cancers in vivo and in vitro.
The antiproliferative activity of escin is investigated in various cell lines such as leukemia, glioma/glioblastoma, cholangiocarcinoma, renal, breast, cervical and lung cancer cells.13 The proapoptotic efficacy of escin has been investigated in many cancers and this ability implies its potency to be an anti-cancer agent. Escin’s apoptosis-triggering mechanisms have been shown to be cell type dependent. Its proapoptotic activity was reported in, human renal,14 human lung adenocarcinoma, C6 glioma,15 and H-Ras transformed 5RP7 cells.16 Studies on the anti-cancer and cytotoxic activities of escin collectively imply the high potency to be an effective anti-cancer agent by triggering apoptosis in different cancer types but the mechanism underlying its anti-cancer and proapoptotic effects on human skin melanoma cells remains unclear. Thus, this study evaluated the possible antiproliferative, proapoptotic, and cytotoxic effects of escin on human skin melanoma (CHL-1) cells.
Materials and methods
Materials
CHL-1 (ATCC® CRL-9446™) cell line was purchased from the American Type Culture Collection (Manassas, USA). Escin, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl-2H-tetrazolium bromide), fetal bovine serum, penicillin–streptomycin, dimethyl sulfoxide (DMSO) were from Sigma-Aldrich (St. Louis, USA), and Dulbecco’s Modified Eagle’s Medium (DMEM) was obtained from GIBCO (Grand Island, USA). Annexin-V and Dead Cell Assay Kit, Bcl-2 Activation Dual Detection Kit, and Oxidative Stress Kit were purchased from (Merck, Millipore, Hayward, California, USA).
MTT assay
In this manner, a stock solution of escin was prepared (in DMSO with 1% final concentration). CHL-1 cells were plated into 96-well plates at a concentration of 1 × 105 cells/well and incubated with different concentrations of escin for 24 h at 37 °C in a humidified incubator of 5% CO2 in the air. After the incubation, 20 μL of MTT solution (5 mg/mL) was added per well and incubated further for 2 h at 37 °C, 5% CO2 incubator. At the end of incubation, the media of each well were changed with 200 μL of dimethyl sulphoxide, and plates were read with a spectrophotometer (HTX-Synergy, Biotek, USA) at a wavelength of 570 nm (n = 3).17
Confocal microscopy
For morphological changes, CHL-1 cells were seeded on coverslips in 6-well plates (3 × 105/well) and treated with IC50 concentration of escin for 24 h at 37 °C, 5% CO2 incubator. After the incubation, cells were washed in PBS and stained with phalloidin and acridine orange for 20 min at room temperature in the dark and imaged with a confocal microscope (Leica SP5-II, The Netherlands).18
Transmission electron microscopy
Ultrastructural changes were investigated by transmission electron microscopy. Briefly, cells treated with IC50 concentration of escin for 24 h were fixed with glutaraldehyde (2.5%, in 0.1 M phosphate buffer (pH 7.4)) and post-fixed in osmium tetroxide (2%). Fixed cells were dehydrated in graded ethanol (70%, 90%, 96%, and absolute alcohol) and then embedded in Epon epoxy. Polymerization of the epoxy was realized with incubation at 60 °C for 48 h. Thin sections (100 nm) were taken using a glass knife by ultramicrotome and were stained with lead citrate and uranyl acetate. Stained sections were imaged under a transmission electron microscope (TEM).18
Phosphatidylserine translocation assay
Annexin-V staining technique was performed to determine the triggered cell death type by escin on CHL-1 cells. For this manner, untreated and escin-treated CHL-1 cells were trypsinized and harvested in separate tubes (100 μL). Annexin-V reagent of 100 μL was added to each tube and all samples were incubated for 15 min at room temperature in the dark following the protocol of the user manual of Muse® Annexin-V and Dead Cell Assay Kit. Cells were analyzed using Muse™ Cell Analyzer (Merck, Millipore, Hayward, California, USA).18
Bcl-2 activation assessment
CHL-1 cells were treated with IC50 concentration for 24 h and untreated CHL-1 cells were harvested by trypsinization and centrifuged at 1200 rpm for 5 min. All samples were washed in PBS and fixed in a fixation buffer (for 5 min) on ice. The fixed samples were washed again in PBS and then permeabilized with permeabilization buffer (for 5 min) on ice. Followed by washing in PBS, 2 × 105 cells were added to tubes and treated with 10 μL of the antibody cocktail and 90 μL assay buffer for 30 min at room temperature (in dark). After the incubation, cells were centrifuged at 1200 rpm for 5 min, washed and resuspended in 200 μL assay buffer per the manufacturer’s instructions for Muse® Bcl-2 Activation Dual Detection Kit. Cells were analyzed by using Muse™ Cell Analyzer (Merck, Millipore, Hayward, California, USA).19
Oxidative stress analysis
CHL-1 control cells and test cells treated with IC50 concentration for 24 h were trypsinized and prepared in separate tubes at a concentration of 1 × 106 in assay buffer according to the manufacturer’s instructions (Muse® Oxidative Stress Kit). Oxidative Stress working solution (190 μL) and 10 μL of cell suspension were mixed and incubated at 37 °C for 30 min. All samples were read on Muse™ Cell Analyzer (Merck, Millipore, Hayward, California, USA).20
Statistics
One-way analysis of variance (ANOVA) and the Tukey’s Honest Significant Difference (HSD) post-test were employed using GraphPad Prism 6.0 for Windows. The data was expressed as mean ± SD and p < 0.05 value was considered statistically significant.
Results
Inhibition of CHL-1 cell growth
CHL-1 cells were incubated with different concentrations of escin for 24 h. The MTT assay findings showed that escin suppressed the growth of CHL-1 cells in a dose-dependent manner (Fig. 1). The IC50 value of escin for this incubation time was found to be 6 μg/mL. The percentages of viable cells decreased significantly at all of the exposed concentrations of escin except for the lowest concentration.
Fig. 1.
Growth inhibition chart of human skin melanoma CHL-1 cells treated with different concentrations of escin for 24 h. (*p < 0.05). The IC50 value of escin on CHL-1 cell for 24 h was determined to be 6 μg/mL.
Confocal microscopic findings
The morphological changes of CHL-1 cells caused by escin are shown in Fig. 2. CHL-1 cells treated with IC50 concentration of escin were damaged with holes in the cytoskeleton, condensed chromatin, and blebbing on membranes. Apoptotic bodies, fragmented nuclei, and pyknotic nuclei were other morphological changes in CHL-1 cells. All these findings suggest that CHL-1 cells underwent apoptosis as a result of escin exposure.
Fig. 2.
Confocal microscopy images of CHL-1 cells. Untreated CHL-1 cells (A and B); Arrow-compact cell morphology, asterisk-compact nuclei. CHL-1 cells treated with escin (C and D); Arrow-holes on cytoskeleton, asterisk-nuclei with condensed chromatin, arrowhead-Membrane blebbings, circle-apoptotic bodies and (E and F); Arrow-Pyknotic nuclei, circle-blebs on cell membrane and arrowhead-fragmented nucleus.
TEM analyses findings of ultrastructural changes in CHL-1 cells
In the ultrastructure of CHL-1 cells treated with escin, we detected damage to membranous organelles of which mitochondria had the highest deformation. The cristae of mitochondria were disintegrated and lost as shown in Fig. 3. Membranes of CHL-1 cells were blebbed after escin treatment. Also, chromatin condensation was remarkably detected in escin-treated CHL-1 cells.
Fig. 3.
Ultrastructure of untreated (A) and escin-treated (B) CHL-1 cells (6,000×). Double headed arrow: Normal cell structure, Arrowhed: Nucleus, arrow: Loss of cristae, asterisk: Chromatin condensation, circle: Membrane blebbings.
Annexin-V findings
The triggered cell death type was detected by the annexin-V technique in escin-treated CHL-1 cells. Apoptotic profiles of human skin melanoma cells showed that the percentages of living CHL-1 cells were 85.71% and 62.28% in control and escin-treated cells, respectively. CHL-1 cells in the early apoptotic stage were 13.71% in untreated cells whereas this percentage in escin-treated cells was detected to be 37.17%. The percentages of late apoptotic cells were found to be nearly unchanged with 0.58% for untreated cells and 0.55% for CHL-1 cells exposed to the IC50 value of escin Fig. 4.
Fig. 4.
Apoptotic profiles human skin melanoma cells. A) Untreated CHL-1 cells; live cells 85.71%, early apoptotic cells 13.71%, late apoptotic cells 0.58%. B) CHL-1 cells treated with IC50 concentration of escin for 24 h; live cells 62.28%, early apoptotic cells 37.17%, late apoptotic cells 0.55%.
Bcl-2 assessment findings of CHL-1 cells
Escin treatment caused suppression in the Bcl-2 signaling pathway with the results for untreated CHL-1 cells a (Fig. 5) which indicates 1.80% for inactivated Bcl-2, 37.70% for activated Bcl-2 and 48.80% for non-expressing CHL-1 cells. Whereas these results were found to be 09.00%, 21.40%, and 71.30% in the cells treated with escin, respectively.
Fig. 5.
Bcl-2 phosphorylation levels of CHL-1 cells. A) Untreated CHL-1 cells; inactivated Bcl-2-1.80%, activated Bcl-2-37.70%, non-expressing cells 48.80%. B) CHL-1 cells treated with IC50 concentration of escin for 24 h; inactivated Bcl-2-09.00%, activated Bcl-2-21.40%, non-expressing cells 71.30%.
Oxidative stress analyses of CHL-1 cells
Reactive oxygen species were detected as 85.83% ROS negative and 0.43% ROS positive cells in control CHL-1 cells (Fig. 6). The ROS profile of CHL-1 cells was changed to 72.77% ROS negative and 9.60% ROS positive cells as a result of escin treatment.
Fig. 6.
ROS profiles of CHL-1 cells. A) Untreated CHL-1 cells; ROS negative cells 85.83%, and ROS positive cells 0.43%. B) CHL-1 cells treated with IC50 concentration of escin for 24 h; ROS negative cells 72.77%, and ROS positive cells 9.60%.
Discussion
Escin is a natural product with many biological activities as anti-inflammatory and antiedemal activities.15,16 Recent studies reported escin to have cytotoxic and anti-cancer activities as well as proapoptotic activity in various cancer cell lines.9,15,16 The experiments on human skin melanoma CHL-1 cells indicated that escin effectively suppressed the proliferation of the cells in a concentration-dependent manner for an application time of 24 h (Fig. 1.). Similarly, escin is reported to inhibit the cell growth on Panc-1, Mia-Paca, COLO357, and P34 pancreatic cancer,9 human lung adenocarcinoma (A549),15 renal cancer14 and rat glioma (C6) cells.15 The significant antiproliferative activity of escin in CHL-1 cells with and low half-maximal inhibitory concentration of 6 μg/mL may be attributed to its efficacy in inducing apoptosis in CHL-1 cells. Morphological and ultrastructural changes like holes on the cytoskeleton, chromatin condensation, blebbing on cell membranes, apoptotic bodies, pyknotic nuclei, fragmented nucleus, and disintegrated mitochondria detected in CHL-1 cells exposed to escin indicated programmed cell death (Figs 2 and 3). Similar alterations in C6 glioma, A549 lung adenocarcinoma, and K562 chronic myeloid leukemia cell lines were reported after exposure to escin and were explained as apoptotic changes.15,21 Based on the detected apoptotic sparks on the ultrastructure and morphology of human skin melanoma CHL-1 cells, the cell death mode triggered by escin was investigated on a flow cytometer by measurement of phosphatidylserine translocation to the outer side of cellular membrane. The results showed that the cell death type induced by escin was apoptosis. The percentage of total apoptotic cells on the CHL-1 cell population treated with escin increased nearly threefold parallelly with the percentage of early apoptotic cells, however the late apoptotic cell population remained constant (Fig. 4.). This finding is supported by previous reports that increased early apoptotic cell population in K562 human chronic myeloid leukaemia cells21 and GIC22 treated with escin. Moreover, studies revealed that escin promotes apoptosis in a time and dose-dependent manner and it caused a significant increase in early apoptosis in incubation time of 24 h while late apoptosis was increased only after 48 h in Jurkat cells.23,24 The unchanged late apoptotic cell population parcentage and significantly increased early apoptotic cell population percentages in CHL-1 cells treated with escin in this study may be explained by the time and dose dependency of escin on promoting apoptosis. Recently, studies have reported the non-specific apoptosis-triggering efficacy of escin on a variety of cancer cell lines.13 Herein, we evaluated the impact of escin on Bcl-2 expression in CHL-1 cells in short-term usage. The results showed that the percentage of Bcl-2 non-expressing CHL-1 cells was increased almost 2-fold by escin application which implies the downregulation of Bcl-2. Furthermore, CHL-1 cell population percentage with activated Bcl-2 was found to be decreased while inactivated Bcl-2 containing CHL-1 cells population percentage was increased upon escin treatment (Fig. 5.). The percentage of inactivated cells indicates that dephosphorylated Bcl-2 expressing CHL-1 cells are part of the Bcl-2 expressing cells, which include both activated and inactivated cells. Briefly, this percentage indicates the dephosphorylated Bcl-2 upon escin treatment and refers to the inactivation of the Bcl-2 signaling pathway. Similarly, studies revealed that escin caused mitochondrial membrane depolarization and downregulation of Bcl-2 expression in human cholangiocarcinoma cells.13,25 Numerous studies provide information suggesting reactive oxygen species and superoxide have been involved in the pathophysiology of various diseases, including cancer.26 The percentage of cells undergoing oxidative stress were determined based on the intracellular superoxide radicals into the CHL-1 cells treated with escin. The study results showed that escin augmented the percentage of ROS positive CHL-1 cells (Fig. 6.). This conclusion is in accordance with our TEM findings, which show that lack of cristae implies mitochondrial malfunction. Other studies revealed that intracellular reactive oxygen species may promote depolarization of the membrane of human cholangiocarcinoma cells.25 Similar results were observed in a study conducted on Jurkat cells by escin administration, which revealed its efficacy on loss of mitochondrial membrane potential, protein content, as well as DNA fragmentation.24 These findings together support the potentials of escin as a cytotoxic, antiproliferative and proapoptotic agent in supressing cells growth in human skin melanoma CHL-1 cells. Briefly, the significant anti-cancer properties of escin indicated in this study, together provide possibilities of designing novel targeted therapeutics for cancer treatment after further investigations on its pharmacokinetic and mechanistic efficacies in different cell lines and in vivo cancer models.
In conclusion, this study assessed the potential anti-cancer efficacy of escin on human skin melanoma CHL-1 cells. Escin was found to exert its anti-cancer activity on CHL-1 cells by inhibiting cell proliferation with the short-time exposure of 24 h. The viability of the escin-treated cells decreased depending on their treatment concentration. Escin was determined to initiate apoptosis in CHL-1 human skin melanoma cells. The morphology and ultrastructure of the escin-treated cells were found to indicate strong apoptosis marks as chromatin condensation, fragmentation of the DNA and nuclei, formation of holes in the cytoskeleton, membrane blebbing, disintegrated mitochondria, cell shrinkage as well as horseshoe-shaped pyknotic nucleus. As a result, escin was shown to have significant potential to be an anti-cancer drug for clinical usage. However, other pharmacokinetic characteristics and deeper mechanisms of action need to be thoroughly studied to develop a novel anti-cancer agent.
Author contributions
The named author was alone without any further contributions. The author planned collected and analyzed the data, wrote, and reviewed the manuscript.
Funding
This study has no funding received for research.
Conflict of interest statement. The author declares no conflicts of interest for this study.
Data availability
Not available.
Ethical approval
This article does not contain any studies with human participants or animals performed by any of the authors.
References
- 1. Organization, W.H . Cancer prevention and control in the context of an integrated approach. Vol. 12. Geneva, Switzerland: World Health Assembly Resolution WHA70; 2017. [Google Scholar]
- 2. International Agency for Research on Cancer. United Kingdom: World cancer factsheet; 2012. https://www.iarc.who.int/wp-content/uploads/2018/07/Global-factsheet-2012-1.pdf.
- 3. Deorukhkar A, Krishnan S, Sethi G, Aggarwal BB. Back to basics: how natural products can provide the basis for new therapeutics. Expert Opin Investig Drugs. 2007:16(11):1753–1773. [DOI] [PubMed] [Google Scholar]
- 4. Ma J, Frank MH. Tumor initiation in human malignant melanoma and potential cancer therapies. Anticancer Agents Med Chem. 2010:10(2):131–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Basmadjian C, Zhao Q, Bentouhami E, Djehal A, Nebigil C, Johnson R, Serova M, De Gramont A, Faivre S, Raymond E. Cancer wars: natural products strike back. Front Chem. 2014:2:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Butler MS. The role of natural product chemistry in drug discovery. J Nat Prod. 2004:67(12):2141–2153. [DOI] [PubMed] [Google Scholar]
- 7. Cragg GM. Paclitaxel (Taxol®): a success story with valuable lessons for natural product drug discovery and development. Med Res Rev. 1998:18(5):315–331. [DOI] [PubMed] [Google Scholar]
- 8. Demain AL, Vaishnav P. Natural products for cancer chemotherapy. Microb Biotechnol. 2011:4(6):687–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rimmon A, Vexler A, Berkovich L, Earon G, Ron I, Lev-Ari S. Escin Chemosensitizes human pancreatic cancer cells and inhibits the nuclear factor-kappaB Signaling pathway. Biochem Res Int. 2013:2013:251752–251759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Gür F, Cengiz M, Kutlu HM, Cengiz BP, Ayhancı A. Molecular docking analyses of Escin as regards cyclophosphamide-induced cardiotoxicity: In vivo and in Silico studies. Toxicol Appl Pharmacol. 2021:411:115386. [DOI] [PubMed] [Google Scholar]
- 11. Cengiz M, Ayhancı A, Kutlu HM. Investigation into the protective effects of Escin on blood cells and cyclophosphamide-induced bone marrow toxicity in rats. Bilecik Şeyh Edebali Üniversitesi Fen Bilimleri Dergisi. 2020:7(2):730–738. [Google Scholar]
- 12. Cengiz M, Kutlu HM, Peker Cengiz B, Ayhancı A. Escin attenuates oxidative damage, apoptosis and lipid peroxidation in a model of cyclophosphamide-induced liver damage. Drug Chem Toxicol. 2022:45(3):1180–1187. [DOI] [PubMed] [Google Scholar]
- 13. Cheong DHJ, Arfuso F, Sethi G, Wang L, Hui KM, Kumar AP, Tran T. Molecular targets and anti-cancer potential of escin. Cancer Lett. 2018:422:1–8. [DOI] [PubMed] [Google Scholar]
- 14. Yuan SY, Cheng CL, Wang SS, Ho HC, Chiu KY, Chen CS, Chen CC, Shiau MY, Ou YC. Escin induces apoptosis in human renal cancer cells through G2/M arrest and reactive oxygen species-modulated mitochondrial pathways. Oncol Rep. 2017:37(2):1002–1010. [DOI] [PubMed] [Google Scholar]
- 15. Çiftçi GA, Işcan A, Kutlu M. Escin reduces cell proliferation and induces apoptosis on glioma and lung adenocarcinoma cell lines. Cytotechnology. 2015:67(5):893–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Güney G, Kutlu HM, İşcan A. The apoptotic effects of Escin in the H-Ras transformed 5RP7 cell line. Phytother Res. 2013:27(6):900–905. [DOI] [PubMed] [Google Scholar]
- 17. Kutlu HM, Sezer CV, Kuş G, Çömlekçi E, İzgördü H. A 5-fluorouracil derivative: Carmofur as a new potent agent for inhibition of human prostate and breast cancer cell lines. Iran J Sci Technol Trans A: Sci. 2022:46(1):13–20. [Google Scholar]
- 18. Kuş G, Kutlu HM, Vejselova D, Çömlekçi E. Comparative study on induction of apoptosis in A549 human lung adenocarcinoma cells by C2 ceramide or Ceranib-2. Pak J Zool. 2017:49(2):591–598. [Google Scholar]
- 19. Palit F, Vejselova Sezer C, Kutlu HM. Efficacy of ceramidase inhibition on human renal cell carcinoma: a cell culture study. Eur Rev Med Pharmacol Sci. 2023:27(5 Suppl):121–129. [DOI] [PubMed] [Google Scholar]
- 20. Cengiz M, Ayhanci A, Akkemik E, Şahin İK, Gür F, Bayrakdar A, Cengiz BP, Musmul A, Gür B. The role of Bax/Bcl-2 and Nrf2-Keap-1 signaling pathways in mediating the protective effect of boric acid on acrylamide-induced acute liver injury in rats. Life Sci. 2022:307:120864. [DOI] [PubMed] [Google Scholar]
- 21. Niu YP, Li LD, Wu LM. Beta-aescin: a potent natural inhibitor of proliferation and inducer of apoptosis in human chronic myeloid leukemia K562 cells in vitro. Leuk Lymphoma. 2008:49(7):1384–1391. [DOI] [PubMed] [Google Scholar]
- 22. Harford-Wright E, Bidère N, Gavard J. β-Escin selectively targets the glioblastoma-initiating cell population and reduces cell viability. Oncotarget. 2016:7(41):66865–66879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Patlolla JMR, Raju J, Swamy MV, Rao CV. β-Escin inhibits colonic aberrant crypt foci formation in rats and regulates the cell cycle growth by inducing p21waf1/cip1 in colon cancer cells. Mol Cancer Ther. 2006:5(6):1459–1466. [DOI] [PubMed] [Google Scholar]
- 24. Mojžišová G, Kello M, Pilátová M, Tomečková V, Vašková J, Vaško L, Bernátová S, Mirossay L, Mojžiš J. Antiproliferative effect of β-escin-an in vitro study. Acta Biochim Pol. 2016:63(1):79–87. [DOI] [PubMed] [Google Scholar]
- 25. Shen DY, Kang JH, Song W, Zhang WQ, Li WG, Zhao Y, Chen QX. Apoptosis of human Cholangiocarcinoma cell lines induced by β-Escin through mitochondrial caspase-dependent pathway. Phytother Res. 2011:25(10):1519–1526. [DOI] [PubMed] [Google Scholar]
- 26. Brieger K, Schiavone S, Miller FJ Jr, Krause K-H. Reactive oxygen species: from health to disease. Swiss Med Wkly. 2012:142:w13659. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not available.






