In vitro and in vivo anticancer effects of marmesin in U937 human leukemia cells are mediated via mitochondrial-mediated apoptosis, cell cycle arrest, and inhibition of cancer cell migration

Retraction in: /10.3892/or.2021.8065

  • Authors:
    • Lin Dong
    • Wen-Wei Xu
    • Hong Li
    • Ke-Hong Bi
  • View Affiliations

  • Published online on: December 11, 2017     https://doi.org/10.3892/or.2017.6147
  • Pages: 597-602
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Leukemia is one of the highly lethal cancers among all pediatric cancers. With limited drug options and the severe side effects associated with the current chemotherapy, there is pressing need to look for new and novel anticancer agents. Against this backdrop, in the present study we evaluated the anticancer activity of a natural coumarin, marmesin against human leukemia cell line U937 and normal human monocytes It was observed that marmesin exhibited an IC50 value of 40 µM and exerted its cytotoxic effects in a dose-dependent manner. However, the cytotoxic effects of marmesin were comparatively lower for the normal human monocytes as evident from the IC50 of 125 µM. Our results indicated that marmesin inhibits colony formation and induces apoptosis dose-dependently. We also investigated the effect of marmesin on the expression of Bax and Bcl-2 proteins. It was observed that marmesin treatment triggered upregulation of Bax and downregulation of Bcl-2 causing significant increase in the Bax/Bcl-2 ratio, marmesin could also induce ROS mediated alterations in mitochondrial membrane potential. Additionally, marmesin induced G2/M cell cycle arrest and significantly inhibited cell migration potential of leukemia cells at the IC50. Remarkably, marmesin prevent tumor growth significantly in vivo at the dosage of 30 mg/kg in vivo. These results strongly indicate that marmesin may prove to be a novel anticancer lead for the management of leukemia.

Introduction

Leukemia is one of the deadly cancers and mainly arises due to malfunction of the bone marrow hematopoietic cells. Among all pediatric cancers, the frequency and lethality of leukemia as a malignant condition ranks first. Leukemia cells have the potential to easily migrate to different parts of the body (1). Chemotherapy, radiotherapy or the combination of the both is currently used for the treatment of leukemia. In some cases, transplantation of bone marrow, as well as palliative care is also essential (2). However, these treatments have several drawbacks which include, but are not limited to, frequent relapse, development of drug resistance and effect on the quality of life of patients (3). Natural products are considered important for the development of new anticancer lead molecules. Owing to their lower side effects they have gained considerable attention in the recent past. Among natural products, coumarins form a large group of secondary metabolites and have been found to have tremendous pharmacological potential ranging from antibacterial, anti-inflammatory to anticancer properties (4,5). Marmesin is an important coumarin which has recently been shown to possess anticancer activity. In the present study, marmesin was evaluated for its anticancer activity against leukemia cancer cell line U937 (human monocytic leukemia cell line) and normal human monocytes. Furthermore, the possible underlying mechanism was determined. Marmesin induced cytotoxicity in leukemia cells by promoting apoptosis through reactive oxygen species (ROS) mediated alterations in mitochondrial membrane potential (∆Ψm) and G2/M cell cycle arrest. Additionally, marmesin inhibited leukemia cancer cell migration at IC50 concentration of marmesin and also inhibited tumor growth in vivo. Taken together, our results indicate that marmesin may prove to be a potential natural anticancer molecule against leukemia.

Materials and methods

Chemicals and other reagents

The chemicals and reagents used in the present study include, marmesin, RPMI-1640, streptomycin, penicillin G, MTT (3-(4, 5-dimethylthiazole-2yl)-2,5-diphenyltetrazolium bromide), DMSO (dimethyl sulfoxide), Rhodamine 123, DCFH-DA (2′,7′-dichlorodihydrofluorescein diacetate) purchased from Sigma. Fetal bovine serum (FBS) was obtained from Gibco and all antibodies, β-actin and Annexin V/PI were purchased from Santa Cruz Biotechnology (Dallas, TX, USA).

Cell culture conditions

Leukemia cancer cell line U937 (human monocytic leukemia cell line) and normal human monocytes were obtained from Cancer Research Institute of Beijing, China, and it was maintained in DMEM and was supplemented with 10% FBS and antibiotics (100 µg/ml streptomycin and 100 U/ml penicillin G) in an incubator at 37°C (5% CO2 and 95% air).

Determination of IC50 by MTT assay

The anti-proliferation effect of marmesin on leukemia cancer cell line U937 and normal human monocytes was evaluated by MTT assay. U937 cells were grown at 1×106 cells per well in 96-well plates for a time period of 12 h and then exposed to 0, 10, 20, 40, 100, 150 and 200 µM marmesin dose for 24 h. To each well, MTT solution (20 µl) was added. To solubilize MTT formazan crystals, 500 µl DMSO was added. ELISA plate reader was used for the determination of optical density.

Colonigenic assay

For colony formation assay, the leukemia cancer cell line at the exponential growth phase were harvested and counted with a hemocytometer. Seeding of the cells was done at 200 cells per well, incubated for a time period of 24 h to allow the cells to attach and then different doses (0, 20, 40 and 80 µM) of marmesin was added. After treatment, the cells were again incubated for 6 days, washing was done with PBS and methanol was used to fix colonies. Afterwards, colonies were stained with crystal violet for about 30 min before being counted under a light microscope.

DAPI staining and detection of apoptosis in leukemia cancer cell line U937

U937 cells at a density of 2×105 cells/well were seeded in 6-well plates and treated with 0, 20, 40 and 80 µM Marmesin for 48 h. The cells were then subjected to DAPI staining. Afterwards, the cell sample was studied and photographs taken by fluorescence microscopy as previously described (6). Annexin V/IP for determination of apoptotic cell populations was determined as previously described (6).

Cell cycle distribution analysis

For cell cycle distribution analysis, the cells were seeded in 6-well plates (2×105 cells/well) and marmesin was administered to the cells at the doses of 0, 20, 40 and 80 µM followed by 24 h of incubation. DMSO was used as a control. For estimation of DNA content, PBS was used to wash the cells and fixed in ethanol at −20°C. This was followed by re-suspension in PBS holding 40 µg/ml PI and, RNase A (0.1 mg/ml) and Triton X-100 (0.1%) for 30 min in the dark at 37°C. Afterwards, analysis was carried out by flow cytometry as previously reported (7,8).

Evaluation of ROS and MMP

U937 cells were seeded at a density of 2×105 cells/well in a 6-well plate and kept for 24 h and treated with 0, 20, 40 and 80 µM marmesin for 24 h at 37°C in 5% CO2 and 95% air. Thereafter cells from all samples were collected, washed two times by PBS and re-suspended in 500 µl of DCFH-DA (10 µM) for ROS estimation and DiOC6 (1 µmol/l) for MMP at 37°C in the dark for 30 min. The samples were then examined instantly using flow cytometer as previously described (8).

Wound healing assay

U937 cells were seeded at a 5×104 cell density in 96-well plates and then allowed to adhere overnight. At confluence, a wound was scratched across each well by WoundMaker device. Afterwards the cells were washed with PBS to remove the detached cells.

Protien expression by western blot analysis

The Marmesin administrated cells were harvested and lysed. The protein concentrations of the lysates were quantified by BCA assay using specific antibodies. β-actin was used as a control. From each sample equal amounts of protein were loaded and separated by electrophoresis on a 12% denaturing SDS gel. Afterwards, the proteins were electroblotted on polyvinylidene difluoride membranes (0.45-µm pore size).

In vivo antitumor effects of marmesin

Twenty severely compromised immunodeficient mice obtained from Cancer Research Institute of Beijing were used in this study. The mice were inoculated intraperitoneally with 1×107 cells in 0.3 ml of PBS. When tumors were substantial, the mice were randomly grouped into two cohorts of 10 mice each. The mice received a daily oral dosage of 30 mg/kg of marmesin for 30 days. Control group received an equal volume of PBS only by gavage. The diameters of the tumor were measured with calipers at 10-day intervals, and tumor volume (mm3) and weight (g) was determined using the standard formula.

Statistical analysis

All experiments were carried out in triplicates and presented as representative images or average values ± SD. The results were considered significant at P<0.01, P<0.001 and P<0.0001.

Results

Cytotoxic potential of marmesin on U937 cell line

The cytotoxic role of marmesin on U937 cells and normal monocytes was detected by treatment of these cells with varied marmesin concentrations. Marmesin displayed the potent anti-proliferative effect against U937 cells with an IC50 40 µM (Fig. 1A). However, the cytotoxic effects of marmesin were comparatively lower on normal human monocytes (IC50 125 µM). In the colony formation assay, we observed that marmesin treated cells showed reduced number of colony formation in a dose-dependent manner (Fig. 1B).

Marmesin induced apoptosis in U937 cells

The apoptotic potential of marmesin on U937 cells was investigated by DAPI staining. Our results indicated that marmesin induced apoptosis dose-dependently as evident from the higher density of apoptotic cells (Fig. 2A). Furthermore, Annexin V/IP staining followed by flow cytometry showed that apoptotic cell population increased from 2.17% in control to 52.55% at 80 µM concentration (Fig. 2B).

Marmesin enhances the Bax/Bcl-2 ratio in U937 cells

Bax and Bcl-2 are key marker proteins for apoptosis and treatment with marmesin resulted in enhanced expression of Bax, (pro-apoptotic protein) and downregulation of Bcl-2 expression (anti-apoptotic protein) leading to incremental increase in the Bax/Bcl-2 ratio in a dose-dependent manner (Fig. 3).

Marmesin induces ROS accretions in U937 cells

The pro-apoptotic potential of marmesin observed through DAPI staining study suggested that marmesin might induce generation of intracellular ROS. Therefore, we calculated the ROS level at varied concentrations of marmesin for 24 h. The results showed that the intracellular ROS levels of treated cells increased up to 211% at 80 µM as compared to untreated cells (Fig. 4A). Our result suggested that marmesin is a potent molecule for activating ROS in U937 cells to trigger apoptosis.

Marmesin lessens the mitochondrial membrane potential (MMP)

ROS generation causes mitochondrial dysfunction. It disrupts the outer mitochondrial potential to release the death-promoting proteins (9). Therefore, we examined whether marmesin reduces the MMP in U937 cells treated with marmesin at varied concentrations (0–80 µM). Treated U937 cells showed a significant reduction in MMP in a dose-dependent manner. The MMP was reduced up to 67% at 80 µM of marmesin as compared to untreated control (Fig. 4B).

Marmesin affects cell cycle distribution

It was observed that the percentage of U937 cells was considerably increased in G2 at the concentrations of 0–80 µM of marmesin causing G2/M arrest. Additionally the populations of U937 cells in G2/M were marginally increased at a dose of 20 µM, reasonably increased at 40 µM, and dramatically increased at 80 µM. This marmesin-induced G2 increase of U937 cancer cells was observed to exhibit a dose-dependent pattern (Fig. 5).

Marmesin affects cell migration in wound healing assay

Further, we examined if marmesin could inhibit the migration of U937 cancer cells at the IC50 concentration by wound healing assay. The results of wound healing assay showed that marmesin reduced the migratory capability of wound healing assay cells, while as in control, the cells show fairly good capacity to migrate, in treatment, the cells showed migration as depicted in Fig. 6.

Marmesin exerts antitumor effects in vivo by reducing tumor size and weight

The antitumor effect of marmesin was also evaluated in vivo. It was observed the administration of marmesin (30 mg/kg) significantly reduced the tumor volume (Fig. 7A) and tumor weight (Fig. 7B) as compared to control group (Fig. 7C).

Discussion

Among all pediatric cancers, the frequency and mortality due to leukemia as a malignant condition ranks first (1). The current treatment options have several associated side effects which severely effect quality of life and hence patient compliance. Plants have proved to be essential sources for development of effective anticancer lead molecules. Of note, more than half of currently used anticancer drugs are from natural products (10). In the current study, we evaluated the anti-cancer activity of marmesin, a natural coumarin, against leukemia U937 cells. The results indicated that the test molecule, marmesin exerted significant anticancer activity in a dose-dependent manner with an IC50 of 40 µM. However, marmesin exerted comparatively lower cytotoxic effects on the normal human monocytes with an IC50 of 125 µM. These results are promising, since lower cytoxicity towards normal cells is considered essential for development of anticancer chemotherapy. Furthermore, marmesin also reduced the colony formation potential of U937 cells dose-dependently. Analysis of apoptotic cells by DAPI and the percentage of apoptotic cell populations by Annexin V/IP staining revealed that marmesin exerted apoptosis in U937 cells in a concentration-dependent manner. This prompted us to study the expression of apoptosis-related proteins by western blotting. We observed that marmesin upregulated Bax expression and at the same time inhibited Bcl-2 expression resulting in increased Bax/Bcl-2 ratio. The increased Bax/Bcl-2 ratio causes activation of caspase-3 and hence apoptosis. As reported previously, many drugs exhibit anti-proliferative effects via induction of apoptosis. For instance, several chemotherapeutic drugs, such as cisplatin, taxol and 5-fluorouracil (814) have been shown to trigger apoptosis and cause DNA damage (15). Further, it was observed that marmesin-induced ROS facilitated reduction in MMP. Therefore, these results suggest that marmesin may induce apoptosis by increasing intracellular ROS and reducing MMP. Our results are in agreement with studies wherein several anticancer drugs have been reported to target cancer cells partly by accretion of high levels of ROS (15). Moreover, mitochondria play a key role in ROS (16).

As previously reported, capsaicin in pancreatic cancer cells reduces MMP and arbitrates oxidative stress ultimately resulting in apoptosis (16,17). Leukemia cells have very high capacity to migrate to other cells (1,2) and in our case marmesin exhibited the capacity to inhibit the cell migration of leukemia cells. These results indicate that marmesin may prevent metastasis of cancer cells in vivo. Given the interesting and promising results in vivo, we also evaluated the antitumor properties of marmesin in vivo. Noteworthy, marmesin at the dosage of 30 mg/kg abrogated the tumor growth. As compared to control the treated group had lower tumor size and weight indicating the anti-leukemic activity of marmesin. Therefore, inhibitory effect of marmesin on leukemia cancer cells may prove crucial in the treatment and management of leukemia. It is important to mention that we evaluated the anticancer activity of marmesin against only one leukemia (U937) cell line, this study will pave the way for further evaluation of marmesin against other cancer cell lines.

Taken together, we conclude that marmesin exhibits significant anticancer activity against leukemia cells by induction of apoptosis and inhibits cell migration which is considered critical for anti-leukemic agents. Furthermore, marmesin also inhibits tumor growth in vivo. Thus, marmesin may prove to be a useful candidate against leukemia and requires further research.

Acknowledgements

This study was funded by Development Plan of Medical and Health Science and Technology in Shandong province (project number: 2016WS0472).

References

1 

Mardiros A, Brown CE, Budde LE, Wang X and Forman SJ: Acute myeloid leukemia therapeutics: CARs in the driver's seat. Oncoimmunology. 2:e272142013. View Article : Google Scholar : PubMed/NCBI

2 

Hoffbrand AV, Moss PAH and Pettit JE: Essential haematology. 5th. Wiley-Blackwell; Malden, MA: 2006

3 

Badura S, Tesanovic T, Pfeifer H, Wystub S, Nijmeijer BA, Liebermann M, Falkenburg JH, Ruthardt M and Ottmann OG: Differential effects of selective inhibitors targeting the PI3K/AKT/mTOR pathway in acute lymphoblastic leukemia. PLoS One. 8:e800702013. View Article : Google Scholar : PubMed/NCBI

4 

Baba SA, Malik AH, Wani ZA, Mohiuddin T, Shah Z, Abbas N and Ashraf N: Phytochemical analysis and antioxidant activity of different tissue types of Crocus sativus and oxidative stress alleviating potential of saffron extract in plants, bacteria, and yeast. S Afr J Bot. 31:80–87. 2015. View Article : Google Scholar

5 

Yadav JP and Panghal M: Balanitesaegyptiaca (L.) Del. (Hingot): A review of its traditional uses, phytochemistry and pharmacological properties. Int J Green Pharm. 4:140–145. 2010. View Article : Google Scholar

6 

Chipuk JE, Bouchier-Hayes L and Green DR: Mitochondrial outer membrane permeabilization during apoptosis: The innocent bystander scenario. Cell Death Differ. 13:1396–1402. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Maitra R, Porter MA, Huang S and Gilmour BP: Inhibition of NFkappaB by the natural product Withaferin A in cellular models of cystic fibrosis inflammation. J Inflamm (Lond). 6:152009. View Article : Google Scholar : PubMed/NCBI

8 

Hissin PJ and Hilf R: A fluorometric method for determination of oxidized and reduced glutathione in tissues. Anal Biochem. 74:214–226. 1976. View Article : Google Scholar : PubMed/NCBI

9 

Azuma M, Tamatani T, Ashida Y, Takashima R, Harada K and Sato M: Cisplatin induces apoptosis in oral squamous carcinoma cells by the mitochondria-mediated but not the NF-kappaB-suppressed pathway. Oral Oncol. 39:282–289. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Amirghofran Z, Bahmani M, Azadmehr A and Javidnia K: Induction of apoptosis in leukemia cell lines by Linum persicum and Euphorbia cheiradenia. J Cancer Res Clin Oncol. 132:427–432. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Yoneda K, Yamamoto T and Osaki T: p53- and p21-independent apoptosis of squamous cell carcinoma cells induced by 5-fluorouracil and radiation. Oral Oncol. 34:529–537. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Abal M, Andreu JM and Barasoain I: Taxanes: Microtubule and centrosome targets, and cell cycle dependent mechanisms of action. Curr Cancer Drug Targets. 3:193–203. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Ferreira CG, Epping M, Kruyt FA and Giaccone G: Apoptosis: Target of cancer therapy. Clin Cancer Res. 8:2024–2034. 2002.PubMed/NCBI

14 

Malaguarnera L: Implications of apoptosis regulators in tumorigenesis. Cancer Metastasis Rev. 23:367–387. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Ding H, Han C, Guo D, Chin YW, Ding Y, Kinghorn AD and D'Ambrosio SM: Selective induction of apoptosis of human oral cancer cell lines by avocado extracts via a ROS-mediated mechanism. Nutr Cancer. 61:348–356. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Kowaltowski AJ, de Souza-Pinto NC, Castilho RF and Vercesi AE: Mitochondria and reactive oxygen species. Free Radic Biol Med. 47:333–343. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Khursheed A, Rather MA and Rashid R: Plant-based natural compounds and herbal extracts as promising apoptotic agents: Their implications for cancer prevention and treatment. Adv Biomed Pharma. 3:225–248. 2016. View Article : Google Scholar

Related Articles

Journal Cover

February-2018
Volume 39 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dong L, Xu W, Li H and Bi K: In vitro and in vivo anticancer effects of marmesin in U937 human leukemia cells are mediated via mitochondrial-mediated apoptosis, cell cycle arrest, and inhibition of cancer cell migration Retraction in /10.3892/or.2021.8065. Oncol Rep 39: 597-602, 2018
APA
Dong, L., Xu, W., Li, H., & Bi, K. (2018). In vitro and in vivo anticancer effects of marmesin in U937 human leukemia cells are mediated via mitochondrial-mediated apoptosis, cell cycle arrest, and inhibition of cancer cell migration Retraction in /10.3892/or.2021.8065. Oncology Reports, 39, 597-602. https://doi.org/10.3892/or.2017.6147
MLA
Dong, L., Xu, W., Li, H., Bi, K."In vitro and in vivo anticancer effects of marmesin in U937 human leukemia cells are mediated via mitochondrial-mediated apoptosis, cell cycle arrest, and inhibition of cancer cell migration Retraction in /10.3892/or.2021.8065". Oncology Reports 39.2 (2018): 597-602.
Chicago
Dong, L., Xu, W., Li, H., Bi, K."In vitro and in vivo anticancer effects of marmesin in U937 human leukemia cells are mediated via mitochondrial-mediated apoptosis, cell cycle arrest, and inhibition of cancer cell migration Retraction in /10.3892/or.2021.8065". Oncology Reports 39, no. 2 (2018): 597-602. https://doi.org/10.3892/or.2017.6147