Skip to main content

Cytotoxic and apoptotic potential of gemini-chrysophanol nanoparticles against human colorectal cancer HCT-116 cell lines

Abstract

Background

Colorectal cancer is among the most common cancers and accounts for nearly 9% of all cancers in the world. Chrysophanol is a naturally occurring anthraquinone exerts a number of pharmacological activities such as anti-inflammation, anti-cancer, anti-bacterial, anti-viral, and anti-oxidant effects. This study aims to produce a novel gemini chrysophanol nanoparticles (Gemini-Chr NPs), and to evaluate its anti-cancer effect on the human colorectal cancer cell lines.

Methods

Gemini-Chr NPs were synthesized through nanoprecipitation method and characterized by dynamic light scattering and scanning electron microscopy, Anti-cancer activities were examined through MTT assay on HCT-116 cancer cells, apoptosis was investigated via Annexin V-FITC/PI dual stain assay. Furthermore, the expression of Bax, Bcl-2 and P53 genes were evaluated using real-time PCR and western blotting assay. 

Results

The average particle diameter of the synthesized Gemini-Chr NPs and zeta potential were recorded as 120 nm and 14.4 mV, respectively. In comparison to the normal cells, the cytotoxicity assay confirmed that Gemini-Chr NPs preferentially killed colorectal cancer cells via induction of apoptosis. Moreover, Gemini-Chr NPs could upregulate the expression of Bax in both cancerous and normal cells (p ≤ 0.05) and decreasing the Bcl-2 expression in only tumor cells (p ≤ 0.01), while the expression of P53 is modulated in tumor cells (p ≤ 0.05).

Conclusions

Gemini surfactants could be considered for efficient delivery and improvement of anti-cancer effect of chrysophanol. Gemini-Chr NPs might have the potential for developing novel therapeutic agent against colorectal cancer.

Peer Review reports

Background

Colorectal carcinoma considers to be a major cause of deaths due to cancer and accounts for approximately 600,000 death cases per year worldwide [1]. Medicinal plants are widely distributed in Kurdistan region of Iraq that are used traditionally for the treatment of different diseases including cancer [2, 3]. Rheum species are known to have medical importance in the treatment of many diseases, Rheum ribes Linn (Polygonaceae) is an Iraqi species of Rheum which is also found in Turkey, Iran, Pakistan, Afghanistan, and Russia. Chrysophanol (1,8-dihydroxy-3-methyl-9,10-anthraquinone), a bioactive molecule in the anthraquinone family which is mainly extracted from R. ribes and other species of Rheum [4,5,6,7]. Previous studies have reported apoptosis induction of Rheum extracts [8, 9], in addition the root methanol extract of R. ribes was recorded with antiproliferative activity on colorectal cancer cell lines, miR-200a/b/c and miR-141 expressions were significant increased (p < 0,05) while Bcl-2, ZEB1, and GATA4 expressions were suppressed [10]. Chrysophanol reported to have different pharmacological activities such as anti-inflammatory [11, 12], neuroprotective [13, 14], antibacterial [7, 15]. Furthermore, chrysophanol reported to have activity against breast and colon cancer cells through NF-κB signaling cascades [16, 17], and through AKT and ERK1/2 signaling pathways it induced cell apoptosis in choriocarcinoma [18], also chrysophanol was found to be used as a cancer metastasis inhibitor in the treatment of colorectal cancer [19]. Product formulations for delivering the naturally derived compounds is important to preserve stability, bioactivity, and bioavailability of the active molecular form, in addition to overcome some other limitations such as preservation and low water solubility, which is the central goal of developing nanoparticles NPs based system. It was reported that chrysophanol NPs significantly minimized tumor size and inhibited tumor growth in BALB/c athymic nude mice. In compared to the free chrysophanol, chrysophanol NPs injection in mice showed higher bioavailability [20]. Gemini surfactants are consisting of two identical conventional surfactants that are linked together by a flexible or rigid spacer, and found to be very effective as drugs delivering system into cells [21]. Therefore, as a suitable way to improve chrysophanol anticancer activity, this research aims to explore the effects of gemini surfactant-chrysophanol (Gemini-Chr) against human colorectal cancer HCT-116 cell lines and to study the possible molecular mechanisms associated with apoptosis induction.

Materials and methods

NPs preparation and characterization

The preparation of Gemini-Chr NPs was carried out by nanoprecipitation method [22]. Briefly, 5 mg of chrysophanol (CAS no.: 481–74-3, Sigma Aldrich, USA) and 100 mg of methoxy-poly (ethylene glycol) urethane Gemini surfactant were dissolved in 5 mL methanol (Merck). After evaporation of methanol by rotatory evaporator, the solution is filtered by 0.2 μm syringe filter, lyophilized, and stored at 4 °C until further use. Chrysophanol was dissolved in phosphate-buffered saline (PBS) before each treatment. Dynamic light scattering (DLS), which is applied for the physicochemical characterization of nanoparticles, was used to measure the zeta potential as well as the hydrodynamic diameter of the Gemini-Chr NPs. Additionally, scanning electron microscopy (SEM) achieved detailed visual image of the Gemini-Chr NPs with high-quality and spatial resolution for characterization and a better understanding of cytotoxicity mechanism [23]. The structural representation of a novel Gemini-Chr NPs is shown in Fig. 1.

Fig. 1
figure 1

The structural representation of a novel Gemini-Chr NPs

Cell culture

In this study, human colorectal cancer cell lines (HCT-116) and Mouse Embryonic Fibroblast normal cells (MEF) were obtained from the National Cell Bank of Iran (Pasteur Institute, Tehran, Iran). DMEM (DMEM/HG; Gibco) with high glucose content culture medium was used to expand the cells. Fetal bovine serum 10% (FBS, Gibco, USA) and Pen-Strep solution 1% (Biochrom GbmH, Berlin, Germany) were added to the basal medium. Cells between passages 3–6 were used for different analyses.

Cell viability assay

The viability of HCT-116 and MEF cells were evaluated after treatment with Gemini surfactant NPs, Chrysophanol, and Gemini-Chr NPs using MTT (3-(4, 5-dimethylthiazole-2-yl)-2, 5- diphenyltetrazolium bromide) method [24]. In brief, an initial number of 1 × 104 cells were suspended in 100 mL of DMEM/HG in each well of 96-well plates containing 10% FBS. Cells were incubated with different concentrations of Gemini surfactant NPs, Chrysophanol, and Gemini-Chr NPs for 24, 48 and 72 h. Supernatants were discarded and replaced with of MTT solution 5 mg/mL. Cells were kept for 3–4 h at 37 ˚C and Dimethylsufoxide (DMSO) solution was added. A microplate reader (Bio-rad, England) was used to read the optical density of each group and expressed as % of the control group.

Flow cytometry analysis

Annexin V-FITC/PI double staining assay was applied to determine apoptosis, using an Annexin V-FITC/PI apoptosis detection kit (BD Pharmingen, San Diego, CA, USA). Gemini-Chr NPs 40 and 60 µM were added to colorectal cancer cells HCT-116 and cultured for 24 h, collected and resuspended in 0.5 mL binding buffer/sample for 5 min. Then 5 μL each of FITC-labeled Annexin-V and PI were added for 15 min at 37 °C in dark. Flow cytometer (Becton Dickinson FACS, Holdrege, NE (Nebraska), USA) was used to analyze the stained samples using FlowJo 7.6.1 Software.

Gene expression analysis

To perform reverse transcription polymerase chain reaction (RT-PCR), total RNA was extracted from both of the control and treated cells using TRIzol™ reagent (Bioneer, Korea) according to the manufacturer’s guideline, picodrop spectrophotometer (Thermo) and 1% agarose gel electrophoresis were used to determine the quality and concentration of RNAs. Easy™cDNA Synthesis Kit (Takara Co., Japan) was used to synthesize complementary DNA (cDNA) by the oligo-dT method following the manufacturer’s instructions. The genes expressions of Bax, Bcl-2 and p53 were done using SYBR Green (Ampliqon, Denmark) RT-PCR analysis using appropriate primers. The primers were designed using Oligo7 software (Table 1). In this study, the total volume for RT-PCR reaction reached 10 μL consisted of 5 μL of SYBR Green PCR master mix, 1 μL of forward and reverse primers, 1 μL of cDNA template, and 3 μL of double distilled water (ddH2O) [24]. Relative gene expression was normalized to β2m as an internal control, and calculated by using the 2−ΔΔCT method [25].

Table 1 Sequences of primer pairs used in PCR

Western blotting

HCT-116 and MEF cell lines were treated with 40 and 60 µM of Gemini-Chr NPs, lysed using 500 μL lysis buffer. Then from each group, 10 μg protein was electrophoresed using 10% SDS-PAGE at 120 V for 45 min and then transferred on to membranes of polyvinylidene difluoride at 120 V for 1.5 h and incubated with Bax (Cat no: sc-7480; Santa Cruz Biotechnology, Inc.), Bcl-2 (Cat no: sc-492; Santa Cruz Biotechnology, Inc.) and p53 (Cat no: sc-126; Santa Cruz Biotechnology, Inc.), at 4 °C for 24 h. The membranes after PBS wash for 3 times were incubated for 1 h at room temperature with appropriate HRP-conjugated secondary antibodies (Cat no: sc-516102 and sc-2357; Santa Cruz Biotechnology, Inc.). The detection of immunoblots were carried out on X-ray films using chemiluminescence ECL solution (Bio-Rad). The relative protein expression was normalized to β-actin (Cat no: sc-47778; Santa Cruz Biotechnology, Inc.) [24].

Statistical analysis

All values represent the mean ± standard deviation of three independent experiments. The one-way analysis of variance (SPSS software v.22.0) was employed to analyze data statistically. Groups considered statistically significant when P < 0.05. GraphPad PRISM version 6.01 software was used for generation of graphs.

Results

Gemini-Chr NPs physicochemical characteristics

Gemini-Chr NPs synthesis and characterization were carried out as described previously, the cluster size distribution obtained from DLS analysis (Fig. 2A) shows the average hydrodynamic diameter to be 120 nm. In addition, zeta potential of Gemini-Chr NPs was calculated to be 14.4 mV which is accepted value for stable dispersion (Fig. 2B). SEM micrograph showed that Gemini-Chr NPs were well separated from each other and spherical in shape (Fig. 2C).

Fig. 2
figure 2

Physical characterization of Gemini-Chr NPs. A Cluster size of Gemini–Chr NPs obtained from DLS measurements. B The zeta potential of Gemini–Chr NPs. C SEM imaging was used to measure mean size of nanoparticles

Gemini-Chr NPs cellular toxicity

MTT assay showed that Gemini–Chr NPs inhibit the proliferation of HCT-116 cancer cell lines in a dose and time dependent manner, (Fig. 3 A). The IC50 of Gemini-Chr NPs on 24, 48, and 72 h in HCT-116 cells was recorded as 60.17, 58.52, and 60.80 µM (p < 0.05), respectively. Furthermore, viability assay showed that Gemini-Chr NPs does not affect normal MEF cells in the IC50 values of cancerous cells. The IC50 values for MEF cells were recorded for concentrations more than 100 µM (Fig. 3 B). Free chrysophanol did not show any toxicity on both HCT-116 and MEFs cells in the concentrations that have been employed for Gemini-Chr NPs (Fig. 3 C and D) and the same thing was recorded for Gemini surfactant NPs on MEFs cells (Fig. 3 E).

Fig. 3
figure 3

The growth-inhibitory effect of Gemini-Chr NPs (A and B), Chrysophanol (C and D), and Gemini surfactant NPs (E) on HCT-116 and MEF cell lines using MTT-based assay. Cells were treated with Gemini-Chr NPs, chrysophanol, and Gemini surfactant NPs in a time- and dose-dependent manner. Data represent mean ± standard deviation of three independent experiments

Apoptosis assay by flow cytometry

Annexin V/FITC assay was employed to study the mode of death in Gemini-Chr NPs treated cells. The results of annexin V-FITC positive cells indicated significantly increased apoptosis of Gemini-Chr NPs treated cells (Fig. 4).

Fig. 4
figure 4

Flow cytometry analysis of mode of cell death in Gemini-Chr NPs treated colorectal cancer cells. Annexin V-FITC/propidium iodide (PI) histograms were obtained from analysis of colorectal cancer HCT-116 cells incubated with 40 and 60 µM of Gemini-Chr NPs for 24 h

Bax, Bcl-2, and P53 genes expression

Real-time PCR was used to further confirm the mode of cell death through the study of apoptotic genes expression, it was revealed that Bax/Bcl-2 expression ratio, a hallmark of apoptosis, was elevated in a dose-dependent manner (Fig. 5A, p ≤ 0.0001). As Fig. 5B illustrates, Bax is over-expressed in both cancerous and normal cells (p ≤ 0.05). However, Bcl-2 as an anti-apoptotic gene is down-regulated in only tumor cells (p ≤ 0.01). Furthermore, the analysis demonstrated that the expression of P53 is significantly modulated in tumor cells rather than normal ones (p ≤ 0.05).

Fig. 5
figure 5

A Expression ratio of Bax/Bcl-2 in non-tumoral MEF and HCT-116 tumor cells. ****: p value ≤ 0.0001; B: Relative expression of Bax, Bcl-2 and P53 as apoptotic genes in both cell lines. *: p value ≤ 0.05, **: p value ≤ 0.01, ***: p value ≤ 0.001

Bax, Bcl-2, and P53 proteins expression

Given that apoptosis inducing agents frequently signal through changes in the expression of Bcl-2 and non Bcl-2 related proteins. Thus, expression of anti-apoptotic Bcl-2 and pro-apoptotic Bax and P53 proteins before and after treatment with Gemini-Chr NPs at 40 and 60 µM was examined in HCT-116 cell lines using western blot analysis (Fig. 6). The results revealed reduction in Bcl-2 expression (p value ≤ 0.0001) while increased expression of Bax and P53 in dose dependent manner (p value ≤ 0.0001).

Fig. 6
figure 6

The expression of Bax, Bcl-2, and P53 in HCT-116 cells treated with 40 and 60 µM of Gemini-Chr NPs for 24 h detected by Western blot analyses. The bands show the expression of proteins in tumor cells. The relative protein expression was normalized to β-actin. Full-length blots/gels are presented in supplementary figure S1. **: p value ≤ 0.01, ***: p value ≤ 0.001, ****: p value ≤ 0.0001

Discussion

Colorectal cancer is the third most diagnosed malignancy with high unacceptable mortality, current chemotherapy drugs which although effective but induce severe side effects to the patients, therefore novel approaches for the treatment of colorectal cancer are needed [26]. The use of plant-based drugs has exhibited incomparable advantages in various fields due to their unique chemical structures and diverse biological and clinical applications [27]. Chrysophanol was reported in several cancer cells to inhibit cell growth and regulate genes and proteins involved in controlling apoptosis, cell invasion, metastasis and cell cycle arrest [28]. The application of NPs as carriers or delivery systems has enhanced the conventional anticancer treatments due to their specificity to cancer cells, that enhance drug efficiency and decrease systemic toxicity [29, 30]. In this study, Gemini-Chr NPs were successfully produced and the results revealed cytotoxicity and cell apoptosis induction in HCT-116 cells. Previous records showed no cytotoxicity of Gemini surfactants on HCT-116 cells [31], and the results of this study showed that Gemini surfactant is a little bit toxic than Gemini-Chr NPs against MEFs cells but non significant and this is probably due to the smaller size for Gemini surfactant that can more easily enter the cells. Gemini surfactant due to its biocompatibility and biodegradability properties can be used for administrating hydrophobic drugs as a safe and cost-efficient system [23], and in comparison with monovalent surfactants offer some advantages such as low critical micellization concentration and high solubilization power [32]. The physicochemical characteristics of NPs are essential for their cellular uptake, and moreover the size and charge of NPs are important properties that could affect their pharmacological properties [33]. Studies showed that NPs with 10–250 nm diameter are convenient for systemic administration due to increase in permeability and overcome physiological barriers [34]. The synthesized Gemini-Chr NPs with 120 nm diameter sphere shaped and 14.4 mV zeta potential giving the possibility to be used for systemic circulation applications with proper dispersion without accumulation, appropriate dimension, and surface charge. The results of MTT assay showed that Gemini-Chr NPs decreases the cell viability of HCT-116 cancer cell lines both in a dose and time dependent, while free chrysophanol did not show a considerable inhibitory effect [35, 36]. Therefore, it was supposed that the activity of chrysophanol is increased significantly which may be due to enhancement of its solubility, uptake, and cytotoxicity using gemini surfactant nanocarriers, further future experiments should be carried out to support this result. Furthermore, Gemini-Chr NPs showed no significant difference in normal MEF cells, indicating its low toxicity. Apoptosis, a programmed cell death, is a tightly regulated process that does not affect bordering cells and is considered as a good target for anticancer treatment [37, 38]. The researchers investigated previously the role of chrysophanol in the modulation of various apoptosis pathways in cancer cells [16, 18, 39]. Flow cytometry analysis revealed that Gemini-Chr NPs induced apoptosis in HCT-116 cells using Annexin V/FITC assay. These data at least show that, similar to previous reports, Gemini-Chr NPs through different pathways induce apoptosis. The Bax/Bcl-2 ratio is considered a critical factor in apoptosis. RT-PCR and western blotting were used to investigate the expression of P53 and its related genes Bcl-2 and Bax before and after treating with Gemini-Chr NPs. The ratio of Bax/Bcl-2 expression is the determining factor for the induction of apoptosis, the results showed that the ratio of Bax/Bcl-2 expression was significantly increased (p ≤ 0.0001) dose-dependently after addition of Gemini-Chr NPs, which confirmed the susceptibility of HCT-116 cells against apoptosis [20]. Furthermore, the analysis demonstrated that the expression of P53 is significantly modulated in HCT-116 tumor cells rather than normal ones. Through control of cell proliferation and apoptosis, the tumor protein P53 has an essential role in tumor development. It was reported that chrysophanol inhibited the proliferation of human mast cells by enhancing P53 protein level [40],in the same line chrysophanol by promoting reactive oxygen species (ROS) has efficacy to induce apoptosis in P53-expressing cancer cells [41]. It was found that increased P53 activity and ROS production occurred in prostate cancer LNCap cells treated with chrysophanol NPs which relies generally on dramatic alterations in mitochondrial morphology during the early stages of apoptotic cell death that involve the network fragmentation and the remodeling of cristae [20]. The increasing in P53 gene activity will activate P21 transcription factors and this prompts binding of cyclin dependent kinase 2 (CDK2) with cyclin E leading to cell cycle stop. Also, the activation of P53 in the cytosol will activate Bax and cause suppression of Bcl-2, and leads to change in permeability of mitochondrial membrane and causes cytochrome c to exit to the cytosol. The reaction of cytochrome c with protease activating factor-1 (APAF-1) through activation of cascade and caspase reaction will triggers DNA-se activation. The DNA-se in turn enters the nucleus leading to fragmentation of the DNA, cleavages of poly ADP-ribose polymerase (PARP), and apoptosis [42]. This research is the first study on evaluation of Gemini-Chr NPs effects against HCT-116 cells growth and apoptosis. However, further molecular analysis needs to be performed to confirm how Gemini-Chr NPs may affect the protein pathways.

Conclusion

Gemini-Chr NPs were successfully synthesized and the cytotoxicity assay suggested their preferential proliferation inhibition and reduced cell viability of HCT-116 colorectal cancer cells in comparison to the normal cells via induction of apoptosis. Additionally, Gemini-Chr NPs upregulated the expression of Bax and p53 but downregulated Bcl-2 expression in HCT-116 cells. Hence, Gemini-Chr NPs might have the potential for developing novel therapeutic agent against colorectal cancer.

Availability of data and materials

The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.

Abbreviations

APAF-1:

Apoptotic protease activating factor-1

CDK2:

Cyclin dependent kinase 2

cDNA:

Complementary DNA

ddH2O:

DDouble distilled water

DLS:

Dynamic light scattering

DMSO:

Dimethylsufoxide

Gemini-Chr NPs:

Gemini chrysophanol nanoparticles

HCT-116:

Human colorectal carcinoma cell lines

MEF:

Mouse embryonic fibroblast normal cells

MTT:

3-(4, 5-Dimethylthiazole-2-yl)-2, 5- diphenyltetrazolium bromide

PARP:

Poly ADP-ribose polymerase

PBS:

Phosphate-buffered saline

PCR:

Polymerase chain reaction

SEM:

Scanning electron microscopy

References

  1. Sung JJY, Ng SC, Chan FKL, Chiu HM, Kim HS, Matsuda T, et al. An updated Asia Pacific Consensus Recommendations on colorectal cancer screening. Gut. 2015;64:121–32.

    Article  CAS  Google Scholar 

  2. Adham AN, Hegazy MEF, Naqishbandi AM, Efferth T. Induction of Apoptosis, Autophagy and Ferroptosis by Thymus vulgaris and Arctium lappa Extract in Leukemia and Multiple Myeloma Cell Lines. Molecules. 2020;25:5016. https://doi.org/10.3390/molecules25215016.

    Article  CAS  Google Scholar 

  3. Adham AN, Naqishbandi AM, Efferth T. Cytotoxicity and apoptosis induction by Fumaria officinalis extracts in leukemia and multiple myeloma cell lines. J Ethnopharmacol. 2021;266:113458.

    Article  CAS  Google Scholar 

  4. Kashiwada Y, Nonaka G-I, Nishioka I, Yamagishi T. Galloyl and hydroxycinnamoylglucoses from rhubarb. Phytochemistry. 1988;27:1473–7. https://doi.org/10.1016/0031-9422(88)80218-8.

    Article  CAS  Google Scholar 

  5. Tosun F, Akyüz KÇ. Anthraquinones and flavonoids from Rheum ribes. Ankara Univ Eczac Fak Derg. 2003;32:31–5.

    Article  CAS  Google Scholar 

  6. Naqishbandi AM, Josefsen K, Pedersen ME, Jger AK. Hypoglycemic activity of Iraqi Rheum ribes root extract. Pharm Biol. 2009;47:380–3.

    Article  CAS  Google Scholar 

  7. Alaadin AM, Al-Khateeb EH, Jäger AK. Antibacterial activity of the Iraqi Rheum ribes root. Pharm Biol. 2007;45:688–90.

    Article  Google Scholar 

  8. Hong J-Y, Chung H-J, Bae SY, Trung TN, Bae K, Lee SK. Induction of Cell Cycle Arrest and Apoptosis by Physcion, an Anthraquinone Isolated From Rhubarb (Rhizomes of Rheum tanguticum), in MDA-MB-231 Human Breast Cancer Cells. J Cancer Prev. 2014;19:273–8.

    Article  Google Scholar 

  9. Lu K, Zhang C, Wu W, Zhou M, Tang Y, Peng Y. Rhubarb extract has a protective role against radiation-induced brain injury and neuronal cell apoptosis. Mol Med Rep. 2015;12:2689–94.

    Article  CAS  Google Scholar 

  10. ÇınarAyan İ, Çetinkaya S, Dursun HG, Süntar İ. Bioactive Compounds of Rheum ribes L. and its Anticancerogenic Effect via Induction of Apoptosis and miR-200 Family Expression in Human Colorectal Cancer Cells. Nutr Cancer. 2020;73:1–16. https://doi.org/10.1080/01635581.2020.1792947.

    Article  CAS  Google Scholar 

  11. Wen Q, Mei L, Ye S, Liu X, Xu Q, Miao J, et al. Chrysophanol demonstrates anti-inflammatory properties in LPS-primed RAW 264.7 macrophages through activating PPAR-γ. Int Immunopharmacol. 2018;56:90–7.

    Article  CAS  Google Scholar 

  12. Jeong H-J, Kim H-Y, Kim H-M. Molecular mechanisms of anti-inflammatory effect of chrysophanol, an active component of AST2017-01 on atopic dermatitis in vitro models. Int Immunopharmacol. 2018;54:238–44.

    Article  CAS  Google Scholar 

  13. Zhao Y, Fang Y, Li J, Duan Y, Zhao H, Gao L, et al. Neuroprotective effects of Chrysophanol against inflammation in middle cerebral artery occlusion mice. Neurosci Lett. 2016;630:16–22.

    Article  CAS  Google Scholar 

  14. Zhao Y, Fang Y, Zhao H, Li J, Duan Y, Shi W, et al. Chrysophanol inhibits endoplasmic reticulum stress in cerebral ischemia and reperfusion mice. Eur J Pharmacol. 2018;818:1–9.

    Article  CAS  Google Scholar 

  15. Orbán-Gyapai O, Liktor-Busa E, Kúsz N, Stefkó D, Urbán E, Hohmann J, et al. Antibacterial screening of Rumex species native to the Carpathian Basin and bioactivity-guided isolation of compounds from Rumex aquaticus. Fitoterapia. 2017;118:101–6.

    Article  Google Scholar 

  16. Ren L, Li Z, Dai C, Zhao D, Wang Y, Ma C, et al. Chrysophanol inhibits proliferation and induces apoptosis through NF-κB/cyclin D1 and NF-κB/Bcl-2 signaling cascade in breast cancer cell lines. Mol Med Rep. 2018;17:4376–82.

    PubMed  PubMed Central  Google Scholar 

  17. Kim SJ, Kim MC, Lee BJ, Park DH, Hong SH, Um JY. Anti-inflammatory activity of chrysophanol through the suppression of NF-κB/caspase-1 activation in vitro and in vivo. Molecules. 2010;15:6436–51.

    Article  CAS  Google Scholar 

  18. Lim W, Yang C, Bazer FW, Song G. Chrysophanol Induces Apoptosis of Choriocarcinoma Through Regulation of ROS and the AKT and ERK1/2 Pathways. J Cell Physiol. 2017;232:331–9.

    Article  CAS  Google Scholar 

  19. Deng M, Xue YJ, Xu LR, Wang QW, Wei J, Ke XQ, et al. Chrysophanol Suppresses Hypoxia-Induced Epithelial-Mesenchymal Transition in Colorectal Cancer Cells. Anat Rec. 2019;302:1561–70.

    Article  CAS  Google Scholar 

  20. Lu L, Li K, Mao YH, Qu H, Yao B, Zhong WW, et al. Gold-chrysophanol nanoparticles suppress human prostate cancer progression through inactivating AKT expression and inducing apoptosis and ROS generation in vitro and in vivo. Int J Oncol. 2017;51:1089–103.

    Article  CAS  Google Scholar 

  21. Menger FM, Littau CA. Gemini surfactants: a new class of self-assembling molecules. J Am Chem Soc. 1993;115:10083–90. https://doi.org/10.1021/ja00075a025.

    Article  CAS  Google Scholar 

  22. Fessi H, Puisieux F, Devissaguet JP, Ammoury N, Benita S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. Int J Pharm. 1989;55:R1-4. https://doi.org/10.1016/0378-5173(89)90281-0.

    Article  CAS  Google Scholar 

  23. Karimpour M, Feizi MAH, Mahdavi M, Krammer B, Verwanger T, Najafi F, et al. Development of curcumin-loaded gemini surfactant nanoparticles: Synthesis, characterization and evaluation of anticancer activity against human breast cancer cell lines. Phytomedicine. 2019;57:183–90.

    Article  CAS  Google Scholar 

  24. Zibaei Z, Babaei E, RezaieNezhadZamani A, Rahbarghazi R, Azeez HJ. Curcumin-enriched Gemini surfactant nanoparticles exhibited tumoricidal effects on human 3D spheroid HT-29 cells in vitro. Cancer Nanotechnol. 2021;12:1–15. https://doi.org/10.1186/s12645-020-00074-4.

    Article  CAS  Google Scholar 

  25. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔCT method. methods. 2001;25:402–8.

    Article  CAS  Google Scholar 

  26. Hernández C, Moreno G, Herrera-R A, Cardona-G W. New hybrids based on curcumin and resveratrol: Synthesis, cytotoxicity and antiproliferative activity against colorectal cancer cells. Molecules. 2021;26:2661.

    Article  Google Scholar 

  27. Yuan H, Ma Q, Ye L, Piao G. The traditional medicine and modern medicine from natural products. Molecules. 2016;21:559.

    Article  Google Scholar 

  28. Xie L, Tang H, Song J, Long J, Zhang L, Li X. Chrysophanol: a review of its pharmacology, toxicity and pharmacokinetics. J Pharm Pharmacol. 2019;71:1475–87.

    Article  CAS  Google Scholar 

  29. Gradishar WJ, Tjulandin S, Davidson N, Shaw H, Desai N, Bhar P, et al. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil–based paclitaxel in women with breast cancer. J Clin Oncol. 2005;23:7794–803.

    Article  CAS  Google Scholar 

  30. Mohd-Zahid MH, Mohamud R, Che Abdullah CA, Lim J, Alem H, Wan Hanaffi WN, et al. Colorectal cancer stem cells: A review of targeted drug delivery by gold nanoparticles. RSC Adv. 2019;10:973–85.

    Article  Google Scholar 

  31. Azeez HJ, Neri F, Hosseinpour Feizi MA, Babaei E. Transcriptome Profiling of HCT-116 Colorectal Cancer Cells with RNA Sequencing Reveals Novel Targets for Polyphenol Nano Curcumin. Molecules. 2022;27:3470.

    Article  CAS  Google Scholar 

  32. Bombelli C, Giansanti L, Luciani P, Mancini G. Gemini surfactant based carriers in gene and drug delivery. Curr Med Chem. 2009;16:171–83.

    Article  CAS  Google Scholar 

  33. Infante MR, Pérez L, Morán MC, Pons R, Mitjans M, Vinardell MP, et al. Biocompatible surfactants from renewable hydrophiles. Eur J lipid Sci Technol. 2010;112:110–21.

    Article  CAS  Google Scholar 

  34. Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9:615–27. https://doi.org/10.1038/nrd2591.

    Article  CAS  PubMed  Google Scholar 

  35. Pandith SA, Hussain A, Bhat WW, Dhar N, Qazi AK, Rana S, et al. Evaluation of anthraquinones from Himalayan rhubarb (Rheum emodi Wall. ex Meissn.) as antiproliferative agents. South African J Bot. 2014;95:1–8. https://doi.org/10.1016/j.sajb.2014.07.012.

    Article  CAS  Google Scholar 

  36. Keser S, Keser F, Karatepe M, Kaygili O, Tekin S, Turkoglu I, et al. Bioactive contents, In vitro antiradical, antimicrobial and cytotoxic properties of rhubarb (Rheum ribes L.) extracts. Nat Prod Res. 2020;34:3353–7. https://doi.org/10.1080/14786419.2018.1560294.

    Article  CAS  PubMed  Google Scholar 

  37. Solano-Gálvez SG, Abadi-Chiriti J, Gutiérrez-Velez L, Rodríguez-Puente E, Konstat-Korzenny E, Álvarez-Hernández D-A, et al. Apoptosis: activation and inhibition in health and disease. Med Sci. 2018;6:54.

    Google Scholar 

  38. Pfeffer CM, Singh ATK. Apoptosis: a target for anticancer therapy. Int J Mol Sci. 2018;19:448.

    Article  Google Scholar 

  39. Lin F-L, Lin C-H, Ho J-D, Yen J-L, Chang H-M, Chiou GCY, et al. The natural retinoprotectant chrysophanol attenuated photoreceptor cell apoptosis in an N-methyl-N-nitrosourea-induced mouse model of retinal degenaration. Sci Rep. 2017;7:1–13.

    Article  Google Scholar 

  40. Han N-R, Kim H-Y, Kang S, Kim MH, Yoon KW, Moon P-D, et al. Chrysophanol, an anthraquinone from AST2017-01, possesses the anti-proliferative effect through increasing p53 protein levels in human mast cells. Inflamm Res. 2019;68:569–79.

    Article  CAS  Google Scholar 

  41. Zhao Y, Chaiswing L, Velez JM, Batinic-Haberle I, Colburn NH, Oberley TD, et al. p53 translocation to mitochondria precedes its nuclear translocation and targets mitochondrial oxidative defense protein-manganese superoxide dismutase. Cancer Res. 2005;65:3745–50.

    Article  CAS  Google Scholar 

  42. Wang I-K, Lin-Shiau S-Y, Lin J-K. Induction of apoptosis by apigenin and related flavonoids through cytochrome c release and activation of caspase-9 and caspase-3 in leukaemia HL-60 cells. Eur J Cancer. 1999;35:1517–25. https://doi.org/10.1016/S0959-8049(99)00168-9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

No funding was obtained for this study.

Author information

Authors and Affiliations

Authors

Contributions

The author, AMN, confirms sole responsibility for the following: conception of the work, analysis and interpretation of data, manuscript preparation, and approved the submitted version.

Corresponding author

Correspondence to Alaadin M. Naqishbandi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not Applicable.

Competing interests

The author declares no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

The uncropped full-length gels and blots for Fig. 6 in HCT-116 cells. Each lane was labelled according to the cropped gels/blots in Fig. 6.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Naqishbandi, A.M. Cytotoxic and apoptotic potential of gemini-chrysophanol nanoparticles against human colorectal cancer HCT-116 cell lines. BMC Pharmacol Toxicol 23, 56 (2022). https://doi.org/10.1186/s40360-022-00597-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40360-022-00597-z

Keywords