EMAN RESEARCH PUBLISHING | <p style="text-align:left">Extract of <em>Urgenia grandiflora</em> inhibited breast cancer cell (MCF-7) proliferation and tumorgenesity</p>
Inflammation Cancer Angiogenesis Biology and Therapeutics | Impact 0.1 (CiteScore) | Online ISSN  2207-872X
RESEARCH ARTICLE   (Open Access)

Extract of Urgenia grandiflora inhibited breast cancer cell (MCF-7) proliferation and tumorgenesity

Ibrahim B. E. El Bashir A,B, Loiy Elsir Ahmed HassanC, Amin M. S. Abdul Majid D, Sakina Yagi A*

+ Author Affiliations

Journal of Angiotherapy 3 (1) 138-146 https://doi.org/10.25163/angiotherapy.3120821273031119

Submitted: 27 July 2019 Revised: 30 October 2019  Published: 03 November 2019 


Abstract

Objective: To investigate the cytotoxicity of Urgenia grandiflora bulbs towards three cancer cell lines; human colorectal carcinoma cell line (HCT 116), human hormone sensitive and invasive breast cancer cell line (MCF-7) and human hormone resistant breast cancer cell line (MDA-MB-231), in addition to endothelial normal EA.hy926 cell line. According to selective antiproliferative effect against MCF-7 breast cancer, U. grandiflora extract was subjected to apoptosis and antitumorgenesity studies on MCF-7 cell line. Methods: Maceration with chloroform: methanol (1:1, v/v) was performed to obtain crude extract. Cytotoxicity was established by colorimetric measurement of cell viability. The effect of the extract on mitochondrial membrane potential, chromatin condensation and nuclear morphology of MCF-7 cells were evaluated using Hoechst 33342 stain. The antitumorgenesity was also determined by evaluation of the ability of extract to suppress the reproductive potential of cell division and colonization after treatment (colongenicity). The effect of extract on migration of tumor cells from their primary growth site to distant locations was evaluated by the wound healing assay. Effect of extract on invasion of matrigel by MCF-7 cells was evaluated using standard methods. Results: U. grandiflora extract showed tumor-specific antiproliferative activity against MCF-7 cells. the extract was also found to exert some toxicity towards the normal cell line EA.hy926 and this could be more likely attributed to its richness in cardiac glycosides. The extract demonstrated programmed cell death features, as it induced cell condensation, membrane flubbing and DNA fragmentation, also it disrupted mitochondria integrity in treated cells. Moreover the extract profoundly inhibited tumorgenisity of MCF-7 via inhibition of cell migration colony formation and cell invasion. Conclusion: U. grandiflora could be a new source of chemotherapeutic for breast cancer.

Key words: Urgenia grandiflora, anticancer, apoptosis, antitumorgenesis in vitro

Public Interest Statement: Ureginea grandiflora has a potential chemotherapeutic effect against breast cancer 

Introduction

GO

Cancer characterized by aggressive uncontrolled growth of malignant cells, which acquired the ability to invade and spread to distance organs (Nath et.al 2013). The International Agency for Research on Cancer (IARC) estimated that about 14.9 million worldwide are cancer patients in 2013, of these 7.7 million were men and 6.9 million were women and by 2035 this number is expected to reach 24 million (Bray et.al 2013). The morbidity of cancer dramatically increased in recent years, every month the Atomic hospital receive 1000 cases, malignancies ranked as the major cause of death in the country (Elebead et al, 2012, Elamin et. Al, 2015). The most frequently mentioned cancers in Sudan were breast 24.8%, leukemia 17%, colon 15%, prostate 14.6%, oral 14.3%, lung13.9%, cervix 10.6%, stomach 9.6% and other scattered cancers (Ahmed et al, 2014).

Conventional treatment approaches have many limitations and require novel therapeutic agents to combat resistance, side effects, and carcinogenesis itself (Foo and Michor, 2014). Natural compounds from plant source can prevent, suppress, or reverse the progression of cancer, which represent excellent option for cancer prevention and treatment.  In fact, medicinal herbs have a long history of use in the treatment of cancer. In review article, Hartwell listed more than 3000 plant species that have reportedly been used in the treatment of cancer (Hartwell, 1982). The search for cancer therapy from plants sources stated in 1950s with the discovery vinca alkaloids (vinblastine & vincristine) and the isolation of the cytotoxic podophyllotoxins (Cragg and, Newmann, 2005; Newman and Cragg, 2007). Saeed et al (2015) evaluated the cytotoxicity of 65 extracts from 35 plants, which are most frequently used in Sudanese traditional medicine for diverse indication including cancer-related symptoms, and they found that Lawsonia inermis, Trigonella foenum-graecum and Ambrosia maritma were the most active crude extracts.

Urginea grandiflora (Family: Hyacinthaceae) is perennial, herbaceous and bulbous plant, distributed in the Red Sea Hills in Eastern Sudan (Andrews FW, 1956). The latex of U. grandiflora obtained from the bulb of the plant used for wound hygiene and wound healing (Sultan et al., 2010). Few studies done on this species, however recently the Urginea maritima (same genus) studied for its cytotoxicity effects among sixty-one Egyptian medicinal plants (El-Seedi et al., 2013). The profound results inspired us to investigate U. grandilora.  In the present study U. grandiflora bulb was screened for its antiproliferative potentials against three cancer cell lines namely; human colorectal carcinoma cell line (HCT 116), human hormone sensitive and invasive breast cancer cell line (MCF 7) and human hormone resistant breast cancer cell line (MDA-MB-231), in addition to normal endothelial EA.hy296 cell line. Selective cytotoxicity was found against MCF-7 cell line and thus the effect of U. grandiflora on apoptosis and antitumorgenisty of this cells line were conducted.

Materials and Methods

GO

2.1 Plant materials

Plant material was collected from eastern Sudan, Erkowit region, in January/2015. Botanical identification and authentication were performed and voucher specimens (No. 2014/UG) have been deposited in Botany Department Herbarium, Faculty of Science, University of Khartoum, Sudan.

2.2 Extract preparation

The bulbs of Ureginea grandiflora were cut into small slices, air dried and grinded to powder.   40 g of plant material was macerated in ratio of organic solvents (chloroform: methanol) (1:1, v/v) (200 mL), at room temperature for 72 h. The filtrate was collected and concentrated at 45ºC under vacuum by rotary evaporator (Buchi, USA) and further dried overnight at 45ºC to obtain (6.952g). Stock solution of the extract was prepared at 10 mg/mL in 100% dimethyl sulfoxide (DMSO). The stock solution as well as DMSO (vehicle) was diluted with cell culture medium, so the highest DMSO concentration exposed to the cells was 0.1 % v/v.

2.3 Preliminary phytochemical screening

The extract was subjected to phytochemical analysis for the detection of terpenoids, alkaloids, saponins (Harborne Jb, 1984), tannins, flavonoids (Sofowora A, 1993), cardiac glycoside and anthraquinones (Trease and Evans, 1989) using standard phytochemical methods.

2.4 Cell viability assay

2.4.1 Cell lines and cell culture

Human Endothelial Cell line (EA.hy 926) purchased from ScienCell, USA. Human colorectal carcinoma cell line (HCT-116), human hormone sensitive and invasive breast cancer line (MCF-7) and human hormone resistant breast cancer cell line (MDA-MB 231) were purchase from ACCT, USA.  All cells maintained in an incubator (Binder, Germany) with temperature 37ºC, 5% CO2 and humidity. EA.hy 926, MDA-MB 231 and MCF-7 were propagated in DMEM (Dulbecco’s Modified Eagle Medium, Sigma, Germany) supplemented with 10% FBS (Foetal Bovine Serum, Sigma, Germany) and 1% PS (Penicillin/ Streptomycin, Sigma, Germany).  HCT-116 was propagated in RPMI-1640 (Sigma, Germany) supplemented with 10% FBS, and 1% PS. Cell culture work was done in sterile conditions using Class II biosafety cabinet (ESCO, USA).

2.4.2 Cell proliferation assay

The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromid) cytotoxicity assay was carried out according to the method described by (Mosmann, 1983) with minor modifications. Cells were seeded at 1.5x104 cells in each well of 96-well plate in 100µL of fresh culture medium and were allowed to attach for overnight. The stock solutions of extracts were diluted in cell culture medium to obtain 100µg/mL and 100µL was added to each well. After 48 h of treatment the medium was aspirated and the cells were exposed to MTT solution prepared at 5mg /mL in sterile PBS added to each well at 10% v/v in respective medium and was incubated at 37°C in 5% CO2 for 3 h. The water insoluble formazan salts was solubilized with 200µL DSMO/well. Absorbance was measured by Magellan – Microplate Reader Software TECAN Group Ltd., Switzerland) at primary wave length of 570 nm and reference wavelength of 620 nm. Each plate contained the samples, negative control and blank. DMSO at less than 1% v/v was used as a negative control. Similarly, tamoxifen with serial concentrations is used as standard reference drug.

2.5 Determination of changes in mitochondrial membrane potential

The changes in mitochondrial membrane potential was detected by Rhodamine 123 staining (Johnson et al., 1980; O'Connor et al., 1988). Briefly, MCF-7 cells (0.5 × 106 cells/mL) were cultured in 24-well plates. After cell attachment, the cells were exposed to new medium with extract (10 and 20µg/mL) or with vehicle (0.1% DMSO). The cells were washed twice with PBS, ?xed with 4% paraformaldehyde for 30 minutes, washed with PBS, and stained for 30 minutes with Hoechst 33258 stain at 10µg/mL and Rhodamine 123 at 5µg/mL simultaneously. The cells were washed twice and observed under the EVOS f1 fluorescent digital microscope (Advanced Microscopy Group, USA). The loss of mitochondrial membrane potential was indicated by the loss of fluorescent intensity in the cells. The images were acquired after 6, and 12 hours. The apoptotic cells were counted in four randomLy selected ?elds per well. The apoptotic index was calculated as the percentage of apoptotic cells compared to the total number of cells.

2.6 Determination of nuclear condensation by Hoechst 33342 stain

The effect of U. grandiflora extract on nuclear fragmentation of MCF-7 was detected by fluorescence microscopy using Hoechst 33258 stain (Oberhammer et al., 1994: Zhang and Xu 2000). Cells were treated with extract (10 and 20µg/mL) and analyzed separately at two different time intervals (12 and 24 hours). 5-furoracel (10µg/mL) used as the positive and 0.1% DMSO as a negative control. The cells were fixed in 4% paraformaldehyde for 30 minutes before staining with Hoechst 33342 stain and incubated for 30 minutes. Nuclear condensation and fragmentation were examined under a fluorescence microscope. Cells with intensely colored, crescent shape, condensed, or fragmented nuclei were considered to be apoptotic. The number of cells with apoptotic morphology was counted in ten randomly selected fields per well. The cells were photographed at 20 × magnifications using an EVOS fluorescent digital microscope (Advanced Microscopy Group, Bothell, USA). The apoptotic index was calculated as the ratio of apoptotic cells to the total number of cells.

2.7 Clonogenicity assay                     

Effect on clonogenicity of MCF-7 cells evaluated by the colony formation assay as previously described by Franken et al (2006). Briefly, cells were harvested and resuspended in fresh DMEM culture medium at density 500 cell/ mL then cells were seeded in 6-well plate at 2 mL/well and incubated at 37ºC and humidified 5% CO2 for 24 hr.  The old media was removed and cells were treated with different concentrations of the extract 2.5,5, 10 and 20 µg/mL in fresh medium. None treated cells were served as negative control and Tamoxifen as positive control. After 48 hr, the medium containing the treatment sample was removed and the cells were washed twice with PBS and a fresh medium was added. Then the cells were incubated for 5 days to allow colonies to form. At the end of incubation, the colonies were fixed in 4% paraformaldehyde for 30 min and stained with 0.25% crystal violet, the colonies were washed to remove the free excess stain and the number of colonies of more than 50 cells was counted using AMG EVOS florescence inverted microscope (4x10 magnification).

The plating efficiency (PE) of untreated cells was determined as follow:

 PE = (the number of colonies formed / the numberof cells seeded) x 100%.

The survival fraction (SF) of treated cells was calculated using the formula:

SF= ((number of colonies formed after treatment)/ (number of seeded cells x PE)) X 100%            

2.8 Migration assay

The assay was carried out as described previously Liang et al. (2007). In brief, MCF-7 were seeded in 6 well plates till the formation of a confluent monolayer after which a wound was created using 200 µL micropipette tip. The detached cells were removed by washing with PBS and the plates were treated with U. grandiflora extract (10µg/ml). The wounds were photo-graphed after 12 and 18 h, and the width of the cell-free wounds was measured using an inverted light microscope supplied with Leica Quin computerized imaging system. Five fields per well were photographed and minimum of 15 readings per field were taken. The results are presented as mean percentage of migration inhibition compared to control.

2.9 Invasion assay

The assay carried out by a miner modification of the Boyden chamber assay using matrigel (Shaw et al., 2006; Baharetha, 2012). Matrigel was used as an artificial basement membrane matrix, this assay is more rigoroustest and it mimics the behaviour of transmigration of cells in vivo. Briefly, 50µL of (1:1 thawed matrigel (10 mg/mL in DMEM medium) was spread into 96-well plate and allowed to solidify by incubation at 37°C in 5% CO2 for 45min. Then MCF-7 cell line were suspended in DMEM medium and immediately seeded at 5×103 cells/well (150 µL/ well). Some of the cells were seeded in DMEM and DMSO only (1%) and used as a negative control, other wells were seeded with DMEM containing the extract (5,10 and 20 µg/mL), then re-incubated in the humidified incubator at 37ºC and 5% CO2 for further 24 hours. Subsequently, the cell culture medium was aspirated carefully to remove the floating and dead cells. After a single wash with PBS the wells were captured using an AMG EVOS florescence inverted microscope (4xmagnification). Quantification of invasion was assessed by counting the number of the invaded cells in the treated wells with comparison to that of the negative control. The number of invading cells was determined, and results are presented as a percentage inhibition relative to the untreated cells. The calculations done as below:

% of inhibition of invasion = (1-(No. of invaded cells(T) /No. of invaded cells (–ve control))) × 100

Where, T: Treated wells with extract.

2.10 Statistical analysis

Each assay was repeated thrice independently with six replicate each. The results were presented as the mean ± standard error of mean (SEM). Fifty percent inhibitory concentration (IC50) values were calculated from concentration-dependent curves using regression analysis in Microsoft Excel 2013. The statistical significance of difference was evaluated by analysis of variance (ANOVA), followed by Tukey’s post hoc test.  A P-value of less than 0.05 was considered significant.

Results

GO

3.1 Phytochemistry

Phytochemical screening showed that the bulb of U. grandiflora is rich in cardiac glycosides. Flavonoids, tannin, alkaloids, saponins, terpenes and sterols were also detected while anthraquinones were absent.

3.2 Effect on cell viability

Extract of U. grandiflora bulb was tested against the three cancerous cells lines MCF7, HCT-116 and MDA-MB 231 and normal human endothelial cell line, EA.hy926 and results are presented in Table 1. The extract displayed obvious evidence of cytotoxicity on concentration dependent manner against MCF7 and EA.hy 926 cells with IC50 values 10.91 and 19.86 µg/mL respectively. The extract was less toxic to the other two cells lines (HCT-116 and MDA-MB 231) where the IC50 value was > 50 µg/mL. Photographs showing the effect of U. grandiflora extract on MCF-7 cells were also taken and the cytotoxic effect of the extract appeared in acute reduction in the number of cells due to inhibition the cell viability. The untreated cells displayed a compact monolayer of growing cancer cells (Fig.1). 

3.3 Effect on mitochondrial membrane potential and nuclear morphology

The effect of U. grandiflora bulb extract at concentrations 5, 10 and 20 µg/mL for 6, 12 and 24 hours on mitochondrial membrane potential of MCF-7 cells was investigated by staining the nucleus with rhodamine 123 stain. Treatment of MCF7 cells with U. grandiflora extract caused, in a dose dependent manner, loss of mitochondrial membrane potential (Fig. 2-a) appearance of nuclear shrinkage, chromatin condensation and nuclear fragmentation indicating signs of early and late apoptosis (Fig. 2-b). At higher concentrations, some cells also revealed the characteristic crescent-shaped nuclei, which is a typical apoptotic nuclear morphology. Apoptotic index of untreated cells was 7.47 ± 0.48, while significant (P = 0.0001) increase of apoptopic index in a concentration dependent manner was observed (Fig. 2-c).

3.4 Inhibition of clonogenicity of MCF-7 cells

The ability of U. grandiflora bulb extract, at 2.5, 5, 10 and 20 µg/mL, to suppress the reproductive potential of cell division and colonization after treatment for 48h was also investigated. Results are presented in Fig. 3. The clonogenicity study on MCF-7 cells indicated that U. grandiflora extract was cytotoxic at all concentrations used, as evidenced by the decrease in the survival fraction (SF). The plating efficiency (PE) of untreated cells was found to be 14.4 ± 0.91% and the SF of the treated cells was 1 ± 0.57 % at 2.5 µg/mL and 0% at 5, 10 and 20 µg/mL. The result was comparable with the standard reference, tamoxifen (10 µg/mL), which inhibited colony formation completely (SF = 0 %.).

3.5 Inhibition of cell migration

Migration of tumor cells from their primary growth site to distant locations is a crucial step in metastatic tumor growth. The effect of U. grandiflora bulb extract on cell migration was evaluated by the wound healing assay. Due to the successful migration of endothelial cells in the untreated group, the wound is almost closed after 15 h, whereas in the U. grandiflora extract treated group, the wound remained open even after 15 h of incubation. The results are presented as average percentage of wound closure which was 67 ± 0.12% and 82 ± 0.68 % after 12 and 15 hours respectively in the untreated cells. The wound closure in treated cells with U. grandiflora extract at concentrations 10 µg/mL was reduced significantly (P <0.001) to 8 ± 1.68% and 24 ± 1.22% after 12 and 15 hours respectively while higher extract concentration (20 µg/mL) reduced it significantly (P <0.001) to 4 ± 1.98 % and 6 ± 0.31 after 12 and 15 hours respectively (Fig 4-a and b).

3.6 Inhibition of cell invasion

The ability of U. grandiflora bulb extract to inhibit MCF7 cells to invade the surrounding tissues was performed on Matrigel matrix. After 24h of treatment with U. grandiflora bulb extract the inhibition in cell invasion was 26.21 ± 5.54%, 50.96 ± 5.12 and 74.42 ± 5.89% at 5,10 and 20 µg/mL respectively (Fig. 5) suggesting the capacity in dose dependent manner of U. grandiflora extract to inhibit MCF7 cells to invade the surrounding tissues.

Discussion

GO

Cancer treatment involves blocking   specific   molecular   mechanisms   in   tumor proliferation by inducing cell death, hindering cell cycle progression or/and inhibiting tumor invasion and angiogenesis (Al Dhaheri et al., 2013). In the present study, U. grandiflora extract showed tumor-specific antiproliferative activity against MCF-7 cells. Analysis of mitochondrial membrane potential and nucleus morphology in MCF-7 cells revealed that U. grandiflora extract induced apoptosis that could be achieved by activating apoptosis triggering signals that cause loss of mitochondrial membrane potential (Fig. 2-a), cell shrinkage, chromatin condensation, and fragmentation (Fig. 2-b) [27]. Furthermore, U. grandiflora extract effectively suppressed, in a dose-dependent manner, MCF-7 cells colony formation (Fig. 3- a and b).

Tumor cells shed daily in blood circulation as part of their movement, and continuously extravasate and proliferate into the secondary sites (Eguchi, 2001). U. grandiflora extract inhibited significantly (P <0.001) cell migration toward the closure of the wound scratch after 12 and 15 h (Fig. 4- a and b) and showed also a capacity to restrict invasion of MCF-7 cells (Fig. 5) into the Matrigel basement indicating its ability to suppress the motility of the breast cancer MCF-7 cells and thus suggesting its antimetastatic potential (Bockhorn,2007; Valster et al., 2005).

Thus, these results suggested that crude extract from bulb of U. grandiflora could be effective to inhibit cell proliferation and attenuate cancer invasiveness and motility. However, the extract was also found to exert some toxicity towards the normal cell line EA.hy926 and this could be more likely attributed to its richness in cardiac glycosides (Newman et al., 2005). Although, cardiac glycosides are known for their clinical uses to treat heart failure for many years (Prassas and Diamandis, 2008), their beneficial effect for the treatment of cancer is controversial. In a review article evaluating the cancer therapeutic potential of cardiac glycosides, Calderón-Montaño et al. (2014), reported some studies demonstrated that cardiac glycosides can inhibit the proliferation of both cancer cells and human nonmalignant at similar very low concentrations and consequently suggested their low potential for cancer therapy. Their toxicity against both cancerous and healthy human cells occur by their ability to inhibit the Na(þ)/K(þ)-pump and subsequent blocking of protein synthesis (Perne A et al., 2009). On the other hand, Babula P et al. (2013) and De S et al. (2016) highlighted in a review article the therapeutic role of cardiac glycosides in cancer treatment. Several alternative mechanisms compatible with their therapeutic use have been suggested particularly in terms of their pharmacodynamic interactions with cytotoxic drugs used in the clinic (Felth et al., 2009). Cardiac glycosides are known to influence the immune response at multiple levels (Kepp et al. 2012). They have antiproliferative activities via their regulation of the cell cycle and were found to selectively inhibited the proliferation of human tumor cells in mouse xenograft models through their effect on the complex mechanisms of cellular signal transduction (Newman et al., 2008; Balunas and Kinghorn 2005). They play an important role in angiogenesis by inhibiting the release of fibroblast growth factor-2 (FGF-2) (potent angiogenesis promoting substance) (De S et al. 2016).

No detailed phytochemical and cytotoxic studies were reported previously for U. grandiflora, however, Proscillaridin A, bufadienolide was isolated from U. maritima (El-Seedi et al., 2013) and was shown to inhibit the DNA topoisomerases I and II and increase the intracellular Ca2+ concentration Winnicka et al. (2010).

In   conclusion, the   present   study   indicated   that U. grandiflora bulb crude extract inhibited significantly the growth, proliferation and invasion of human breast cancer (MCF-7) cells and induced apoptosis in a time-and-dose dependent manner. To the best of our knowledge this is the first detailed study that address the antitumor property of U. grandiflora. It is well known that; plant extracts comprise mixtures of complex metabolite which in turn exert their action on different levels and through several mechanisms [40]. Thus, isolation and determination of bioactive molecule(s) responsible for inhibitory potential against breast cancer cells from bulb crude extract of U. grandiflora and their mode of action are warranted to evaluate their potential for cancer therapy.

Conflict of interest statement

GO

 The authors have no conflict of interest.

Acknowledgements

GO

Authors would like to acknowledge Prof. Maha Kordof ani (Botany Department, Faculty of Science, University of Khartoum) for the identification of the plant.

Authors contributions

GO

I.B.E.B. run the in vitro assay, L.E.A.H. assisted with experimental protocol and drafted the paper, A.M.S. designed the study, S.A.Y. drafted and revised the article.

References


Ahmed HG, Eltom FM, Doumi MA, Eltybe MM, Mahmoud TA, Ebnoof EA, Salih RA. (2014). Burden of Cancer in North Sudan: A community-based Survey; Egypt. Acad J Biolog Sci; 6(2): 55- 63.
https://doi.org/10.21608/eajbsc.2014.16030
 
Almeida CA., 2010. Cancer: basic science and clinical aspects. Wiley-Blackwell, London.
 
Al Dhaheri Y, Attoub S, Arafat K, AbuQamar S, Viallet J, Saleh A, Al Agha H, Eid A, Iratn R. (2013). Anti-metastatic and anti-tumor growth effects of Origanum majorana on highly metastatic human breast cancer cells: inhibition of NFκB signaling and reduction of nitric oxide production. PLoS ONE; 8(7): e68808.
https://doi.org/10.1371/journal.pone.0068808
PMid:23874773 PMCid:PMC3707896
 
Andrews FW. (1956). The Flowering Plants of the Sudan: Volume III (Compositae-Gramineae) T. Buncle & Co., Ltd,
 
Babula P, Masarik M, Adam V, Provaznik I, Kizek R. (2013). From Na+ /K+-ATPase and cardiac glycosides to cytotoxicity and cancer treatment. Anti-Cancer Agents Med Chem; 13(7): 1069 - 1087.
https://doi.org/10.2174/18715206113139990304
PMid:23537048
 
Baharetha HM. (2012). The use of Nigella sativa Linn. supercritical carbon dioxide extract for breast cancer herapy by targeting the angiogenesis cascade. Msc Thesis, USM.
 
Balunas MJ, Kinghorn ADI. (2005). Drug discovery from medicinal plants. Life Sci; 78: 431-441.
https://doi.org/10.1016/j.lfs.2005.09.012
PMid:16198377
 
Bockhorn M, Jain RK, Munn LL. (2007). Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol; 8: 444-448.
https://doi.org/10.1016/S1470-2045(07)70140-7
 
Bray F, Ren JS, Masuyer E, Ferlay J. (2013). Global estimates of cancer prevalence for 27 sites in the adult population in 2008. Int J Cancer; 132(5): 1133-45.
https://doi.org/10.1002/ijc.27711
PMid:22752881
 
Calderón-Montaño JM, Burgos-Morón E, Luis Orta M, Maldonado-Navas D, García-Domínguez I, López-Lázaro M. (2014). Evaluating the cancer therapeutic potential of cardiac glycosides. Bio Med Res Int; Article ID 794930, 9 pages.
https://doi.org/10.1155/2014/794930
PMid:24895612 PMCid:PMC4033509
 
Cragg GM, Newmann DJ. (2005). Plants as source of anticancer agents. J. Ethnopharmacol; 100: 72-79.
https://doi.org/10.1016/j.jep.2005.05.011
PMid:16009521
 
De S, Banerjee S, Babu MN, Lakhmi BM, Babu TMS. (2016). Review on Cardiac glycosides in cancer research and cancer therapy. Indo Amer J Pharmaceut Res; 6(05).
 
Elebead FM, Hamid A, Hilmi HS, Galal H. (2012). Mapping cancer disease using geographical information system (GIS) in Gezira State-Sudan. J Community Health; 37(4): 830-9.
https://doi.org/10.1007/s10900-011-9517-9
PMid:22227773
 
Elamin A, Ibrahim ME, Abuidris D, Mohamed KEH, Mohammed SI. (2015). Part I: cancer in Sudan-burden, distribution, and trends breast, gynecological, and prostate cancers. Cancer Medicine; 4(3): 447-456.
https://doi.org/10.1002/cam4.378
PMid:25641872 PMCid:PMC4380970
 
Eguchi K. (2001). Apoptosis in autoimmune diseases. Intern Med; 40: 275-284.
https://doi.org/10.2169/internalmedicine.40.275
PMid:11334384
 
El-Seedi HR, Burman R, Mansour A, Turki Z, Boulos L, Gullbo J, Göransson U. 2013The traditional medical uses and cytotoxic activities of sixty-one Egyptian plants: Discovery of an active cardiac glycoside from Urginea maritima. J. Ethnopharmacol; 145: 746-757.
https://doi.org/10.1016/j.jep.2012.12.007
PMid:23228916
 
Felth J, Rickardson L, Rosén J, Wickstroõm M, Fryknas M, Lindskog M, Bohlin L, Gullbo J. (2009). Cytotoxic effects of cardiac glycosides in colon cancer cells, alone and in combination with standard chemotherapeutic drugs. J Nat Prod; 72: 1969-1974.
https://doi.org/10.1021/np900210m
PMid:19894733
 
Franken NP, Rodermond H, Stap J, Haveman J, Bree C. Clonogenic assay of cells in vitro. Nat Protoc 2006; 1(5): 2315-2319.
https://doi.org/10.1038/nprot.2006.339
PMid:17406473
 
Foo J, Michor F. Evolution of acquired resistance to anti-cancer therapy. (2014). J Theor Biol; 355: 10-20
https://doi.org/10.1016/j.jtbi.2014.02.025
PMid:24681298 PMCid:PMC4058397
 
Harborne Jb. (1984). Phytochemical Methods. 2nd edition, Springer; p 37-99.
https://doi.org/10.1007/978-94-009-5570-7_2
 
Hartwell JL. (1982). Plants Used Against Cancer. Quarterman, Lawrence, MA.
 
Johnson L, Walsh M, Chen L. (1980). Localization of mitochondria in living cells with rhodamine 123. Proc Natl Acad Sci; 77: 990-994.
https://doi.org/10.1073/pnas.77.2.990
PMid:6965798 PMCid:PMC348409
 
Kepp O, Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, Sukkurwala A, Michaud M, Galluzzi L, Zitvogel L, Kroemer G. (2012). Anticancer activity of cardiac glycosides: At the frontier between cell-autonomous and immunological effects. Oncoimmunology; 1(9): 1640-1642.
https://doi.org/10.4161/onci.21684
PMid:23264921 PMCid:PMC3525630
 
Liang CC, Park A, Guan JL. (2007). In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc; 2(2): 329-333.
https://doi.org/10.1016/j.tiv.2007.04.005
PMid:17560072
 
Lombardi VRM, Carrera I, Cacabelos R. (2017). In vitro screening for cytotoxic activity of herbal extracts. Evid Based Complementary Altern Med; Article ID 2675631: 8 pages.
https://doi.org/10.1155/2017/2675631
PMid:28386288 PMCid:PMC5366791
 
Mosmann T. (1983). Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunological Methods; 65: 55-63.
https://doi.org/10.1016/0022-1759(83)90303-4
 
Nath R, Roy S, De B, Choudhury MD. (2013). Anticancer and antioxidant activity of croton: A review. Int J Pharm Sci; 5: 63-70.
 
Newman DJ, Cragg GM. (2007). Natural products as sources of new drugs over the last 25 years. J Nat Prod; 70:461-477.
https://doi.org/10.1021/np068054v
PMid:17309302
 
Newman RA, Yang P, Pawlus AD, Block KI. (2008). Cardiac glycosides as novel cancer therapeutic agents. Mol Interv; 8(1): 36-49.
https://doi.org/10.1124/mi.8.1.8
PMid:18332483
 
Oberhammer FA, Hochegger K, Froschl G, Tiefenbacher R, Pavelka M. (1994). Chromatin condensation during apoptosis is accompanied by degradation of lamin A+B, without enhanced activation of Cdc2 kinase. J Cell Biol; 126: 827-37.
https://doi.org/10.1083/jcb.126.4.827
PMid:8051209 PMCid:PMC2120132
 
O'Connor JE, Vargas JL, Kimler BF, Hernandez-Yago J, Grisolia S. (1988). Use rhodamine 123 to investigate alterations in mitochondrial activity in isolated mouse liver mitochondria. Biochem Biophys Res Commun; 151: 568-73.
https://doi.org/10.1016/0006-291X(88)90632-8
 
Perne A, Muellner MK, Steinrueck M, et al. (2009). Cardiac glycosides induce cell death in human cells by inhibiting general protein synthesis. PloS ONE; 4(12): ArticleIDe8292.
https://doi.org/10.1371/journal.pone.0008292
PMid:20016840 PMCid:PMC2788214
 
Prassas I, Diamandis EP. (2008). Novel therapeutic applications of cardiac glycosides. Nat Rev Drug Discov; 7(11): 926-935.
https://doi.org/10.1038/nrd2682
PMid:18948999
 
Saeed MEM, Abdelgadir H, Sugimoto Y, Khalid HE, Efferth T. (2015) Cytotoxicity of 35 medicinal plants from Sudan towards sensitive and multidrug-resistant cancer cells. J. Ethnopharmacol; 174: 644-658.
https://doi.org/10.1016/j.jep.2015.07.005
PMid:26165828
 
Shaw LM. (2005). 'Tumor cell invasion assays', Methods in molecular biology; 97-105.
 
Sofowora A. (1993). Recent trends in research into African medicinal plants. J Ethnopharmacol; 38(2): 197-208.
https://doi.org/10.1016/0378-8741(93)90017-Y
 
Sultan HAS, Abu Elreish BI, Yagi SM. (2010). Anatomical and phytochemical studies of the leaves and roots of Urginea grandiflora Bak. and Pancratium tortuosum Herbert. Ethnobot Leaflets; 14: 826-35.
 
Trease GE, Evans WC. (1989). Pharmacognosy. 11th ed. London: Brailliar Tiridel Can Macmillan Publishers; p. 60-75.
 
Valster A, Tran NL, Nakada M, Berens ME, Chan AY, Symons M. (2005). Cell migration and invasion assays. Methods; 37: 208-215.
https://doi.org/10.1016/j.ymeth.2005.08.001
PMid:16288884
 
Winnicka K, Bielawski K, Bielawska A, Miltyk W. (2010). Dual effects of ouabain, digoxin and proscillaridin A on the regulation of apoptosis in human fibroblasts. Nat Prod Res; 24(3): 274-285.
https://doi.org/10.1080/14786410902991878
PMid:20140806
 
Zhang JH, Xu M. (2000). DNA Fragmentation in apoptosis. J Cell Res; 10: 205-211.
https://doi.org/10.1038/sj.cr.7290049
PMid:11032172

Committee on Publication Ethics

PDF
Abstract
Export Citation

View Dimensions


View Plumx


View Altmetric




Save
0
Citation
687
View

Share