Abstract
Aim:
To determine the antiproliferative and cytotoxic activities of Geranium and Erodium species against human cancer and noncancer cell lines, respectively.
Methods:
Twenty-one species of Geranium and Erodium were extracted and screened against cancerous and noncancerous human cell lines.
Results:
In a dose-response manner, G. glaberrimum, G. asphodeloides, E. brandianum and E. leucanthum were able, with variable potency, to inhibit cellular proliferation. Except for E. brandianum, all extracts induced cellular autophagy in tumor cells with similar levels to that of rapamycin; but, only E. brandianum induced cellular apoptosis, likely through Bcl2 and BAX protein expressions.
Discussion:
This is the first study to report the potential antiproliferative effects of ethanol extracts of several Geraniaceae species.
Keywords: : anticancer, antiproliferative, breast cancer, cytotoxicity, Erodium, Geraniaceae, Geranium
Lay abstract
Several plant species of the genera Geranium and Erodium were extracted and screened against five tumor cell lines for their potential antitumor activities. Out of the tested species, four showed potential antitumor action against all the tumor cells used. Interestingly, they appeared to achieve their antitumor activities by inducing important cellular defense mechanisms called autophagy and apoptosis. The current work is the first to test the antiproliferative potential of these species against different tumor cells.
Anticancer drugs are considered the first line of treatment for some types of cancer [1]. There are several anticancer drugs with different origins and mechanisms of action available to treat certain types of cancers, but the majority of these drugs cause serious side effects. For instance, alkylating agents, topoisomerase inhibitors and antimicrotubule agents directly target DNA replication and cell division in cancer and normal cells, leading to serious side effects [2–4]. Therefore, there is a need to develop new anticancer drugs with fewer side effects. Medicinal plants have always been a valuable source of therapeutic agents and drug leads, including anticancer drugs. It was estimated that 60% of all chemotherapeutic agents approved by the FDA, including vincristine, vinblastine and taxol, trace their origin to natural products or their derivatives [5]. Natural-based drugs are those that contain pharmacologically active ingredients derived from biological or mineral sources and intended for use in the diagnosis, cure, mitigation and prevention of disease [6].
Geranium L. (Geraniaceae) is a large genus comprising almost 400 plant species [7] and was traditionally used for the treatment of various illnesses, including diabetes, hemostasis, diarrhea, cough, whooping cough, kidney pain, tonsillitis and gastrointestinal ailments [8]. However, some studies suggest that the Geranium species is potentially cytotoxic [9–12]. Another major plant genus belonging to Geraniaceae is Erodium L'Herit which consists of 74 species predominantly found in the Mediterranean region [13]. Many of the member species were reportedly used in folk medicines in various countries to treat infections, wounds, hepatitis, constipation, coagulation, hemorrhoid, toothache, flu and diabetes [14]. In addition, some Erodium species were shown to have cytotoxic activity against MT-4, MCF7 and Madin–Darby bovine kidney cells [15,16]. The two genera are also known to have rich phytochemical compositions, particularly polyphenols [12]. The latter was shown to affect cellular apoptosis and senescence processes, which are considered vital processes for cancer proliferation; however, their effectiveness appeared to be cell-type dependent [17].
The ethnopharmacological, bioactivity and phytochemical data on these species demonstrate that the two genera, namely Geranium and Erodium, represent a potentially rich source of biologically active components. While some of their biological activities have been reported, their antiproliferative potential has not been explored. Thus, it is hypothesized that screening of Geranium and Erodium species against tumor cells might shed new light on the therapeutic potential of these genera. The current study aims to evaluate the potential in vitro antiproliferative activity and cytotoxicity of ethanol extracts from Geranium and Erodium genera using cell-based assays against several cancer and noncancerous cell lines, respectively. In vitro cell-based assays have been extensively used as a tool for in vitro drug screening, including anticancer screening, as they enable the rapid determination of potential antiproliferative activity. Therefore, screening plant extracts using a reliable drug screening methodology may result in the identification of effective, less toxic and novel anticancer drug leads.
Methods
Plant materials & extraction procedure
Plant materials were extracted using the maceration method with ethanol (95%), which is a general extraction technique for medicinal plants. Briefly, powdered plant materials were placed with a solvent in a stoppered container at room temperature for at least 3 days. When the dissolution of the soluble phytochemicals was complete, the liquid part was separated by filtration [18]. In total, 21 ethanol extracts were prepared from Erodium absinthoides: Willd. subsp. absinthoides (four locations), E. amanum Boiss. and Kotschy, E. birandianum Ilarslan and Yurdakulol, E. cedrorum Schott and Kotschy ssp. cedrorum, E. ciconium (L.) L'Herit., E. cicutarium (L.) L'Herit., E. gruinum (L.) L'Herit., E. hendrikii Alpinar, E. leucanthum Boiss., E. malacoides (L.) L'Herit., E. pelargoniiflorum Boiss. and Heldr., E. somanum H. Peşmen as well as Geranium asphodeloides Burm., G. lasiopus Boiss. and Heldr., Geranium glaberrimum Boiss. and Heldr (two locations), G. lucidum L., G. purpureum Vill. and G. tuberosum L. (two locations). Locations and dates of collection as well as herbarium numbers for all plant species screened are shown in Table 1. The plant samples were collected, identified and preserved in a private herbarium collection at the Scientific Analysis Technological Application and Research Center (Usak University, Turkey). The aerial parts of the collected plant samples were cut into small pieces and dried at room temperature. The dried pieces were ground to a fine powder using a mechanical grinder. The dried and powdered aerial parts of the plants were extracted with 95% ethanol (EtOH) over a 5-day period with casual manual stirring. The EtOH phases of each macerate were independently filtered using Whatman filter paper (150 mm) and concentrated under vacuum in a rotary evaporator (Büchi, Switzerland) to yield the crude extracts. The extracts were preserved in a deep freezer until the experiments.
Table 1. . Collection locations, dates and herbarium numbers for the screened plants.
| Species | Endemism (E) | Collection location, date and herbarium number |
|---|---|---|
| Erodium absinthoides Willd. subsp. absinthoides (Trautv.) Davis (no. 1) | E | Ankara province, Ayaş Mountain, June 2014, A Kahraman 1824 |
| Erodium absinthoides Willd. subsp. armenum (Trautv.) Davis (no. 2) | – | Erzincan province, North of Sipikör Mountain, July 2014, 2472 m, A Kahraman 1872 |
| Erodium absinthoides Willd. subsp. armenum (Trautv.) Davis (no. 3) | – | Erzincan province, Sipikör pass on Sipikör Mountain, July 2014, A Kahraman 1873 (a) |
| Erodium absinthoides Willd. subsp. haradjianii (Davis) Davis (no. 4) | E | Osmaniye province, Düziçi, Düldül Mountain, September 2014, T Dirmenci |
| Erodium amanum Boiss. and Kotschy | E | Hatay province, Belen, Çobandere, June 2014, A. Kahraman 1854 |
| Erodium birandianum Ilarslan and Yurdakulol | E | Kastamonu province, Devrekani, Yaraligöz Mountain, July 2014, A Kahraman 1859, 1830 m |
| Erodium cedrorum Schott and Kotschy subsp. cedrorum | E | Adana province, Aladag, Kicak village, Tahtali upland, June 2014, A Kahraman 1855 |
| Erodium ciconium (L.) L'Herit. | – | Isparta province, Isparta to Antalya, 10 km to Elsazi, May 2014, A Kahraman 1768 |
| Erodium cicutarium (L.) L'Herit. | – | Kütahya province, 15 km from Gediz to Kütahya, May 2014, A Kahraman 1810 |
| Erodium gruinum (L.) L'Herit. | – | Antalya province, Akseki to Manavgat, Murtiçi, May 2014, A Kahraman 1786 |
| Erodium hendrikii Alpinar | E | Gümüşhane province, Yagmurdere, July 2014, A. Kahraman 1893 |
| Erodium leucanthum Boiss. | E | Denizli province, Babadag, above Saçmagedigi, May 2014, A Kahraman 1815 |
| Erodium malacoides (L.) L'Herit. | – | Antalya province, Akseki to Manavgat, Murtiçi, May 2014, A Kahraman 1783 |
| Erodium pelargoniiflorum Boiss. & Heldr. | E | Isparta province, Sütçüler, Kuzca village, 1 km to Tota, June 2014, A Kahraman 1825 |
| Erodium somanum H. Peşmen | E | Manisa province, Soma, above Kızılören village, Sifa Mountain, May 2014, A Kahraman 1798 |
| Geranium asphodeloides Burm. | – | Kastamonu province, Ilgaz Mountain National Park, July, 2014, A Kahraman 1858 |
| Geranium lasiopus Boiss. and Heldr. | E | Antalya province, Akseki Güzelsu, June 2016, A Kahraman 2397 |
| Geranium glaberrimum Boiss. and Heldr. (no. 1) | Konya province, Ibradi-Derebucak road, near Derebucak, 23.04.2016, A Kahraman 2308 | |
| Geranium glaberrimum Boiss. and Heldr. (no. 2) | E | Antalya province, Akseki, Sadiklar village, May 2014, A Kahraman 1781 |
| Geranium lucidum L. | – | Konya province, Ibradi-Derebucak road, near Derebucak, 23.04.2016, A Kahraman 2309 |
| Geranium purpureum Vill. | – | Antalya province, Köprülü canyon, 22.04.2016, A Kahraman 2298 |
| Geranium tuberosum L. (no. 1) | – | Antalya province, Elmalı, near Uzungeriş hill, June 2014, A Kahraman 1838 |
| Geranium tuberosum L. (no. 2) | – | Antalya province, Akseki to Beyşehir, 54 km to Beyşehir, May 2014, A Kahraman 1777 |
subsp.: Subspecies.
In vitro screening
Cell lines
Breast cancer cells MCF-7 (a gift from the Tulchinsky lab), noncancerous fibroblastic cells NIH/3T3 (a gift from Carmen Birchmeier's lab), muscle Ewing's sarcoma cells A-673 (ATCC® CRL-1598™, USA), epithelial cervical carcinoma DoTc2 4510 (ATCC® CRL-7920™, USA), hepatocellular carcinoma HUH-7 (gifted by the National Laboratory, Astana) and malignant melanoma A-375 (ATCC® CRL-1619™, USA) cell lines were cultured in normal glucose Dulbecco's modified Eagle's medium (DMEM, Sigma, MO, USA) supplemented with fetal bovine serum (FBS; 10% [v/v]; Sigma) and streptomycin and gentamycin (1% [v/v]; Sigma). Cells were incubated in a humidified atmosphere at 37°C and 5% CO2.
Antiproliferative & cytotoxicity assays
Cellular proliferation assay
Cellular proliferation was determined using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method [19]. Briefly, cells were seeded at 1 × 104 cells/mL in 96-well microtiter plates in minimum essential medium with FBS and incubated overnight for attachment. A final volume of 100 μl of serial log dilutions of different test compounds (blind screening) was added to each well in triplicate (final compound concentrations ranged from 20 ng/ml to 200 μg/ml). Cells were incubated in the presence of different investigational drugs at 5% CO2 at 37°C for 24 h. Cells were then treated with MTT (Sigma, MO, USA) and incubated for 4 h. After the incubation period, formazan crystals were dissolved by MTT solubilization solution (Sigma) and the absorbance was measured at 570 nm using a 96-well imaging reader (Cytation™ 5, Bio-Tek Instruments, Inc., USA). The cytotoxicity index was determined using the untreated cells as the negative control. The percentage of antiproliferative activity (in cancer cells) and cytotoxicity (in noncancerous cells) were calculated according to the method described by Florento et al. using the treated reading/negative control (nontreated); cell viability (%) = [(Asample at 570 nm/Acontrol at 570 nm)] × 100 [20].
Therapeutic index
The therapeutic index (TI) for each of the tested extracts was determined by dividing the extract's IC50 by its CC50, which was determined using dose-response and dose-cytotoxicity graphs, respectively. Briefly, the extract concentration that inhibited cancer cell proliferation by 50% (IC50) was divided by the extract concentration that reduced the cellular viability of noncancerous cells by 50% (CC50). Both values were determined by plotting data points for a range of drug concentrations and calculating the values using regression analysis.
Autophagy assay
Cellular autophagy was measured using an Autophagy Assay kit (Abcam, UK) according to the manufacturer's guidelines. Cells were cultured in a 96-well black plate with a clear bottom at a density of 1 × 104 cells/ml, then treated with the extracts at different concentrations (final concentrations 20 ng/ml to 200 μg/ml), and incubated at 5% CO2 at 37°C for 24 h. Following the incubation period, the medium was removed from the cells and 100 μl of the autophagosome detection reagent working solution were added to each well (samples and controls), and then incubated at 37°C with 5% CO2 for 30 min. Cells were washed three times by gently adding 100 μl of wash buffer to each well and was then carefully removed to prevent dislodging the cells. Fluorescence intensity was then measured using an imaging reader (CytationTM5, Bio-Tek Instruments, Inc., USA) at λex360/λem520 nm.
Apoptotic protein expression using western blot
MCF-7 cells were incubated for 24 h and then treated with doxorubicin (1 uM) alone or cotreated with 200 µg/ml of E. birandianum. After 48 h of treatment, the cells were harvested and lysed using loading dye containing 1% Triton X-100, 50 mM Tris-Cl (pH: 6.8), 100 mM NaCl 1% SDS, 10% glycerol, β-mercaptoethanol and bromophenol blue. Protein concentrations were determined and equal quantities of protein (40–50 μg) were loaded on 12% SDS-PAGE and transferred to polyvinylidene fluoride (PVDF) membranes. The membranes were incubated in a blocking buffer (Tris-buffered saline [TBS] buffer containing 0.1% Tween 20 [Sigma] and 5% nonfat dry milk) for 1 h at room temperature to block nonspecific binding. The membranes were then incubated with primary antibodies, anti-β actin, anti-BAX and anti-BCL2 at 4°C overnight. The membranes were washed in TBS, and incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h at room temperature and then detected using an enhanced chemiluminescence (ECL) detection system. For band intensity comparisons and quantification, NIH ImageJ software was used (http://rsb.info.nih.gov/ij/).
Statistical analysis
All data are expressed as mean ± standard error of the mean unless otherwise stated. Data were analyzed using one-way analysis of variance (ANOVA) and Student's t-tests. Results with p-values less than 0.05 were considered statistically significant.
Results
Antiproliferative & cytotoxic activity
A total of 21 plant extracts were evaluated in a blind manner for their potential antiproliferative activity against five cell lines: human breast cancer cell lines (MCF7), A673 muscle Ewing's sarcoma, 4510 epithelial cervical carcinoma, HuH7 hepatocellular carcinoma and A375 malignant melanoma. The results showed that 10 extracts displayed antiproliferative activity at high concentrations (200 μg/ml). Interestingly, four of the tested extracts belonging to G. asphodeloides, G. glaberrimum (no. 2), E. leucanthum and E. birandianum inhibited cellular proliferation in a dose-response manner with different potency against different cancer cell lines (Figure 1A–E). However, in order to determine the cytotoxicity of the extracts and to confirm their antiproliferative specificity, the extracts were screened again using noncancerous NIH/3T3 cells. Of the extracts screened, E. birandianum was shown to be significantly less toxic than the others (Figure 2).
Figure 1. . Inhibition of cellular proliferation.

Inhibition of cellular proliferation following 24 hours treatment with serial concentrations of doxycycline, Geranium glaberrimum, Erodium brandianum, Erodium leucanthum and Geranium asphodeloides. The inhibition of cellular proliferation was measured in (A) MCF-7, (B) A-673, (C) Do-TC2 4510, (D) HUH-7 and (E) A-375 cells. The results shown are the average of three independent experiments.
Figure 2. . Cellular toxicity.
The potential cellular toxicity of four active plant extracts and doxycycline were measured using noncancerous fibroblastic cell line, NIH/3T3. Cells were treated with serial concentrations of each of the extracts for 24 hours. The results shown are the average of three independent experiments.
Therapeutic index
The IC50 and CC50 values for each of the tested extracts were determined by the dose-response curves shown in Figures 1A–E & 2, respectively. As shown in Table 2, E. birandianum had the highest TI in all tested cell lines, which was much higher than doxycycline (positive control; Table 2). The TI for E. birandianum extract was found to be significantly higher than the other three extracts in all of the treated cells, but significantly greater than the positive control in HUH7 (α <0.05) and A375 (α = 0.04) cells only.
Table 2. . Therapeutic indices for the tested extracts.
| Cell line | G. glaberrimum (no. 2) | G. asphodelioides | E. leucanthum | E. birandianum | Doxycycline |
|---|---|---|---|---|---|
| MCF-7 | 1.9 | 2.2 | 2.8 | 6.0 | 4 |
| A673 | 1.2 | 1.3 | 1.0 | 6.5 | 3.5 |
| 4510 | 1.8 | 1.0 | 1.5 | 5.5 | 3.8 |
| HUH7 | – | 0.9 | 1.6 | 5.9 | 2.7 |
| A375 | 1.9 | 0.9 | 1.5 | 5.2 | 3.3 |
Autophagy assay
To determine the potential antiproliferative mechanisms of the extracts, the concentration of IC50 for each extract was selected and screened in MCF7 cells, since it is the most sensitive cell line for these extracts as determined by the TI values. The active extracts, except for E. birandianum, induced autophagy in MCF7 cells with variable potency, but not in the noncancerous fibroblastic cell lines (NIH/3T3; Figure 3). At a final concentration of 2 μg/ml, the extract from E. leucanthum induced a 5-fold increase in autophagy compared with nontreated cells; the G. asphodeloides extract induced an approximately 4.2-fold increase compared with nontreated cells; and G. glaberrimum (no. 2) induced autophagy at approximately 3.8-times that of the nontreated cells. However, no significant difference was observed between the autophagy inducer (rapamycin) and the extracts of G. glaberrimum (no. 2; α = 0.06), G. asphodeloides (α = 0.18) and E. leucanthum (α = 0.65; Figure 3).
Figure 3. . Induction of autophagy.
The autophagy induction abilities of the four tested active plant extracts were compared with the positive control, rapamycin. MCF-7 cells were treated with each of the plant extracts for 24 h. The results show a fold increase over the relevant negative control (no treatment).
*Statistically significant difference (p < 0.05). The results shown are the average of three independent experiments.
Apoptosis protein expression
The results of the assessment of cellular apoptosis showed that treatment with doxorubicin alone or combined with E. birandianum, but not E. birandianum alone, significantly reduced the expression of Bcl-2 (antiapoptotic) cells compared with the control (Figure 4A & B). İnterestingly, E. birandianum alone or combined with doxorubicine resulted in a significant increase in the expression levels of the proapoptotic BAX protein compared with the control (Figure 4A & C).
Figure 4. . Protein Expressions.

(A) Levels of expression of BCL-2 and BAX proteins. (B) In comparison to negative control, treatment with doxorubicin or combined with E. birandianum, significantly reduced the expression of BCL-2. (C) Also, E. birandianum or in combination with doxorubicin significantly increased the expression levels of BAX protein compared to the control.
Discussion
Natural products have been the most productive sources of leads for the development of new drugs, especially anticancer agents. Many of today's therapeutic agents owe their existence to natural products, with more than half of all new drug approvals still tracing their structural origins to a natural product [21,22]. Newman et al. claimed that 55% of chemotherapeutic drugs approved between 1981 and 2010 were derived from or based on natural products [23]. Thus, natural products will remain one of the most important sources of therapeutic entities for the treatment of various diseases.
In the current study, in vitro screening using the cell-based MTT assay was applied to a number of ethanol extracts from the aerial parts of a number of Erodium and Geranium species against several cancer cell lines. MTT assay is a reliable colorimetric assay commonly used for chemotherapeutic drug screening [20]. The assay measures cellular metabolic activity as an indicator of cell viability, proliferation and cytotoxicity. In principle, metabolically active cells reduce yellow tetrazolium salt (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to purple formazan crystals; the darker the solution the greater the number of viable cells [24,25]. Interestingly, the results demonstrated that the EtOH extracts of G. glaberrimum, G. asphodeloides, E. brandianum, and E. leucanthum were able, with variable potency, to inhibit cellular proliferation of breast cancer cells (MCF-7), muscle Ewing's sarcoma cells (A-673), epithelial cervical carcinoma (DoTc2 4510), hepatocellular carcinoma (HUH-7) and malignant melanoma cells (A-375).
The tested extracts were previously reported to display different biological activities. For example, some of the chemical constituents of G. glaberrimum (no. 2), which is endemic to the southwestern region of Turkey [26], were reported to have antiplasmodial and cholinesterase inhibitory activities [27], while the chemical constituents isolated from E. brandianum were reported to possess antioxidant and antimicrobial activities [28]. E. leucanthum was shown to have antimitotic activity [29]. Interestingly, G. asphodeloides, the plant extract with the highest TI, was reported to possess several biological activities including antimicrobial and antioxidant activity [30]. Compared with doxycycline used as the positive control, the extracts showed potent antiproliferative activity against the screened cell lines with G. asphodeloides having the most potent therapeutic activity, even higher than that of doxycycline. The extracts showed a similar level of cytotoxicity in nontumor fibroblastic cells (NIH/3T3) to that of the positive control in the concentration range between 20 ng/ml and 20 μg/ml. However, at 200 μg/ml, E. brandianum extract was significantly less toxic than the control, whereas G. glaberrimum (no. 2) had a similar CC50 to the control, but both extracts (G. asphodeloides and E. leucanthum) displayed high cellular toxicity. While all of the extracts displayed potential antiproliferative activity comparable to the control, the extract of E. brandianum exhibited potent antiproliferative activity and low cytotoxicity, resulting in a high TI. Therefore, E. brandianum has significant potential for further studies as a potential therapeutic entity not only as chemotherapy but against other diseases that were reportedly treated by other members within the genus Erodium L.
Recent phytochemistry analyses of E. birandianum, by the authors' group as part of a recent and unpublished PhD thesis led to the identification of a number of phenolics, such as quercetin, as the major component. Other identified components in this plant include gallic acid, caffeic acid, quinic acid, syringic acid, 3,4-dihydroxybenzoic acid, catechin, epicatechin, rutin, luteolin and apigenin (unpublished data by Baki Kekilli) [31]. Therefore, the antiproliferative activity of E. birandianum extract shown in the current results is likely associated with one or many of the identified components. The major identified flavonoid, quercetin, was shown to be cytotoxic to MCF-7 cells and, in combination with mycophenolic acid, was in a synergistic manner able to inhibit breast cancer cell growth [32–34]. In a recent study by Yamada et al. [35], quercetin was reported to successfully suppress hepatocyte growth factor (HGF) or transforming growth factor-α (TGF-α) in HuH7 cell lines, which was also confirmed by others [36]. Therefore, based on the current finding that quercetin is the major phenolic component in E. birandianum, and the recently reported antiproliferative effects of quercetin, we speculate that quercetin is one of the main components responsible for the antiproliferative effect of this particular species.
There are several mechanisms by which these extracts can achieve antiproliferative activity, including autophagy, a cellular process for degrading and eliminating damaged organelles and misfolded proteins that functions in adaptation to cellular development, death and tumor suppression [37]. The autophagic process is controlled by different proteins, including the mammalian target of rapamycin (mTOR), which is associated with cell proliferation and cancer progression [38]. Notably, G. asphodeloides, E. leucanthum and G. glaberrimum (no. 2), but not E. brandianum, through as-yet-unknown mechanisms, induced autophagy in MCF7 cells with a similar potency to that of rapamycin (the positive control). Consequently, it can safely be assumed that autophagy initiation is likely to be one of the antiproliferative mechanisms of the active extracts, which is a common mechanism seen in the PI3K/Akt/mTOR axis-inhibiting anticancer drugs [39].
Another interesting mechanism that was tested is the induction of the apoptotic pathway, which is thought to be an effective method for the treatment of cancer, as the cell numbers are dependent upon the extent of cell proliferation and death [35]. However, there are several mechanisms that cancer cells use to evade apoptosis, including the overexpression of the antiapoptotic Bcl2 family of proteins, a protein family that regulates the mitochondrial cell-intrinsic apoptotic pathway [36,37]. The Bcl2 protein binds to proapoptotic members, such as BAX, a protein that induces cell death in tumor cells, to prevent pore formation and cytochrome c release [38]. Cell treatment with doxorubicin alone or combined with E. birandianum resulted in markedly lower levels of Bcl2 protein expression compared with the control. Treatment with E. brandianum alone also resulted in a statistically insignificant reduction in the protein expression level of Bcl2 compared with the control. In contrast, cell treatment with doxorubicin alone, E. brandianum alone or combined increased the expression level of BAX protein compared with the control. These data show that cellular treatment with E. brandianum can alter the ratio of Bcl2 and BAX proteins, which is an important step in deciphering the potential antiproliferative effects of E. brandianum. Thus, it is safe to suggest that E. brandianum is an inducer of cellular apoptosis through changes in the protein expression ratio of Bcl2 and BAX.
The results in the present study describe the potential antiproliferative activities of 4 out of 21 tested plant extracts from several Geranium and Erodium species. The data demonstrated remarkable antiproliferative activity of the four plant extracts against five different cancer cell lines with overall TI values comparable to those of the positive control, doxycycline. Although the exact mechanisms of action of these extracts are not fully determined, all of the extracts, except E. brandianum, seemed to induce cellular autophagy in tumor cells with similar levels to that of rapamycin. Also, E. brandianum appears to induce cellular apoptosis mediated through Bcl2 and BAX protein expressions.
Conclusion
The current study is a proof of concept that medicinal plants can provide a substantial number of drug lead candidates with various biological activities. This is the first study to report the potential anticancer effects of ethanol extracts of the Geraniaceae species. Only 4 of the 21 screened species, G. asphodeloides, G. glaberrimum (no. 2), E. leucanthum and E. birandianum displayed antiproliferative activities in a dose-response manner against different cancer cell lines. While the majority of the screened species induced cellular autophagy in tumor cells, only E. brandianum caused apoptosis. Further studies are required to identify the bioactive components of the extracts that showed antiproliferative effects and to determine their mechanisms of action.
Summary points.
There is a need for effective and less toxic anticancer drugs.
Medicinal plants represent an invaluable source of therapeutic agents, including anticancer drugs.
Geraniaceae is a plant genus that comprises more than 400 species.
A number of plant species have therapeutic uses and have been used in folk medicine in different regions.
Five cancerous cell lines were used to test the antiproliferative potential of the ethanol-extracted species.
Only four of the extracts showed potential antiproliferative activity in a dose-response manner.
The noncancerous cell line NIH/3T3 was used to determine the cytotoxic effects.
E. birandianum was shown to have a higher therapeutic index than doxycycline.
In combination with doxorubicin, E. birandianum significantly reduced the expression of Bcl-2 and increased the expression level of BAX, compared with control.
Natural products are a good source of drug leads.
In vitro screening of Erodium and Geranium species using MTT assay against five different cell lines showed potential antiproliferative activities.
Four of the extracts showed low toxicity compared with the control.
The polyphenol quercetin is a major component of E. birandianum, which may be responsible for the observed antiproliferative potential.
The potential antiproliferative mechanism of action of G. asphodeloides, E. leucanthum and G. glaberrimum (no. 2) might be facilitated by autophagy.
The potential mechanism of action of E. birandianum is likely to be mediated through its apoptotic potential, as shown by its ability to decrease Bcl-2 protein expression and increase the BAX protein expression compared with the control.
Footnotes
Author contributions
S Sergazy and A Vetrova performed experiments and wrote the first draft; IE Orhan, FSS Deniz and A Kahraman performed plant collection and ethanol extraction; J Zhang performed western blotting and manuscript writing; M Aljofan provided funding, experimental design and manuscript writing.
Financial & competing interests disclosure
This work was supported through Social Policy Research Grant; PI-M.A provided by Nazarbayev University School of Medicine (NUSOM) and by the Nazarbayev University Collaborative Research Program 2021-2023 (Award no. OPCRP2021006). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
Ethical conduct of research
The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations.
Open access
This work is licensed under the Creative Commons Attribution 4.0 License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
References
Papers of special note have been highlighted as: • of interest; •• of considerable interest
- 1.Koch S, Mayer F, Honecker F, Schittenhelm M, Bokemeyer C. Efficacy of cytotoxic agents used in the treatment of testicular germ cell tumours under normoxic and hypoxic conditions in vitro. Brit. J. Cancer 89(11), 2133–2139 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]; •The article describes the efficacy of different agents and the impact of the environment on their actions.
- 2.Hemminki K, Kallama S. Reactions of nitrogen mustards with DNA. IARC Sci. Publ. (78), 55–70 (1986). [PubMed] [Google Scholar]
- 3.Rowinsky EK, Onetto N, Canetta RM, Arbuck SG. Taxol: the first of the taxanes, an important new class of antitumor agents. Semin. Oncol. 19(6), 646–662 (1992). [PubMed] [Google Scholar]
- 4.Asai A, Koseki J, Konno M et al. Drug discovery of anticancer drugs targeting methylenetetrahydrofolate dehydrogenase 2. Heliyon 4(12), e01021 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Katz L, Baltz RH. Natural product discovery: past, present, and future. J. Ind. Microbiol. Biot. 43(2–3), 155–176 (2016). [DOI] [PubMed] [Google Scholar]
- 6.Alkhamaisah S, Aljofan M. Prevalence of use and reported side effects of herbal medicine among adults in Saudi Arabia. Complement Ther. Med. 48, 102255 (2020). [DOI] [PubMed] [Google Scholar]
- 7.Aedo C, Garmendia FM, Pando F. World checklist of Geranium L. (Geraniaceae). Anales Jard. Bot. 56, 211–252 (1998). [Google Scholar]
- 8.Calzada F, Cerda-Garcia-Rojas CM, Meckes M, Cedillo-Rivera R, Bye R, Mata R. Geranins A and B, new antiprotozoal A-type proanthocyanidins from Geranium niveum. J. Nat. Prod. 62(5), 705–709 (1999). [DOI] [PubMed] [Google Scholar]
- 9.Adam M, Elhassan GOM, Yagi S et al. In vitro antioxidant and cytotoxic activities of 18 plants from the Erkowit region, Eastern Sudan. Nat. Product Bioprosp. 8(2), 97–105 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sim MO, Jang JH, Lee HE, Jung HK, Cho HW. Antioxidant effects of Geranium nepalense ethanol extract on H2O2-induced cytotoxicity in H9c2, SH-SY5Y, BEAS-2B, and HEK293. Food Sci. Biotechnol. 26(4), 1045–1053 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Venskutonis PR, Dedonyte V, Lazutka J et al. A preliminary assessment of singlet oxygen scavenging, cytotoxic and genotoxic properties of Geranium macrorrhizum extracts. Acta Biochim. Pol. 57(2), 157–163 (2010). [PubMed] [Google Scholar]; ••
- 12.Abbasi BA, Iqbal J, Ahmad R et al. Bioactivities of Geranium wallichianum leaf extracts conjugated with zinc oxide nanoparticles. Biomolecules 10(1), 38 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fiz O, Vargas P, Alarcon ML, Aldasoro JJ. Phylogenetic relationships and evolution in Erodium (Geraniaceae) based on trnL-trnF sequences. Syst. Bot. 31(4), 739–763 (2006). [Google Scholar]
- 14.Munekata PES, Alcantara C, Collado MC et al. Ethnopharmacology, phytochemistry and biological activity of Erodium species: a review. Food Res. Int. 126, 108659 (2019). [DOI] [PubMed] [Google Scholar]
- 15.Esmaeili S, Naghibi F, Mosaddegh M, Sahranavard S, Ghafari S, Abdullah NR. Screening of antiplasmodial properties among some traditionally used Iranian plants. J. Ethnopharmacol. 121(3), 400–404 (2009). [DOI] [PubMed] [Google Scholar]
- 16.Ma CM, Nakamura N, Miyashiro H et al. Screening of Chinese and Mongolian herbal drugs for anti-human immunodeficiency virus type 1 (HIV-1) activity. Phytother. Res. 16(2), 186–189 (2002). [DOI] [PubMed] [Google Scholar]
- 17.Serpil Ö, Eda B, Hilal K, Hafize SV, Sevinç Y. Quercetin-mediated apoptosis and cellular senescence in human colon cancer. Anti-Cancer Agents Med. Chem. 20(11), 1387–1396 (2020). [DOI] [PubMed] [Google Scholar]
- 18.Handa S, Khanuja S, Longo G, Rakesh D. Extraction technologies for medicinal and aromatic plants. Earth, Environmental and Marine Sciences and Technologies, United Nations Industrial Development Organization and the International Centre for Science and High Technology, Trieste, Italy: (2008). [Google Scholar]
- 19.Park JG, Kramer BS, Steinberg SM et al. Chemosensitivity testing of human colorectal carcinoma cell lines using a tetrazolium-based colorimetric assay. Cancer Res. 47(22), 5875–5879 (1987). [PubMed] [Google Scholar]
- 20.Florento L, Matias R, Tuano E, Santiago K, Dela Cruz F, Tuazon A. Comparison of cytotoxic activity of anticancer drugs against various human tumor cell lines using in vitro cell-based approach. Int. J. Biomed. Sci. 8(1), 76–80 (2012). [PMC free article] [PubMed] [Google Scholar]
- 21.Stratton CF, Newman DJ, Tan DS. Cheminformatic comparison of approved drugs from natural product versus synthetic origins. Bioorg. Med. Chem. Lett. 25(21), 4802–4807 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Aljofan M, Netter HJ, Aljarbou AN et al. Anti-hepatitis B activity of isoquinoline alkaloids of plant origin. Arch. Virol. 159(5), 1119–1128 (2014). [DOI] [PubMed] [Google Scholar]; •
- 23.Newman DJ, Cragg GM. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 75(3), 311–335 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; ••
- 24.Vistica DT, Skehan P, Scudiero D, Monks A, Pittman A, Boyd MR. Tetrazolium-based assays for cellular viability: a critical examination of selected parameters affecting formazan production. Cancer Res. 51(10), 2515–2520 (1991). [PubMed] [Google Scholar]
- 25.Maehara Y, Anai H, Tamada R, Sugimachi K. The ATP assay is more sensitive than the succinate dehydrogenase inhibition test for predicting cell viability. Eur. J. Cancer Clin. Oncol. 23(3), 273–276 (1987). [DOI] [PubMed] [Google Scholar]
- 26.Walters RWaG. Geranium Glaberrimum Geraniaceae. Blackwell Publishing Ltd, Oxford, UK: (2007). [Google Scholar]
- 27.Lenta BN, Devkota KP, Ngouela S et al. Anti-plasmodial and cholinesterase inhibiting activities of some constituents of Psorospermum glaberrimum. Chem. Pharm. Bull. 56(2), 222–226 (2008). [DOI] [PubMed] [Google Scholar]
- 28.Hamza G, Emna BH, Yeddes W, Dhouafli Z, Moufida TS, El Akrem H. Chemical composition, antimicrobial and antioxidant activities data of three plants from Tunisia region: erodium glaucophyllum, Erodium hirtum and Erodium guttatum. Data Brief 19, 2352–2355 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Robles AJ, Peng JN, Hartley RM, Lee B, Mooberry SL. Melampodium leucanthum, a source of cytotoxic sesquiterpenes with antimitotic activities. J. Nat. Prod. 78(3), 388–395 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Söhretoglu D, Ekizoglu M, Özalp M, Sakar K. Free radical scavenging and antimicrobial activities of three Geranium species growing in Turkey. Fabad J. Pharm. Sci. 32(2), 59–63 (2007). [Google Scholar]
- 31.BK E. Pharmacognostic Researches on Establishing New Xanthine Oxidase Inhibitors of Herbal Origin. In: Department of Pharmacognosy-Institute of Health Sciences. (Ed.). Gazi University, Ankara, Turkey: (2019). [Google Scholar]
- 32.Khan MS, Tabrez S, Al-Okail M et al. Non-enzymatic glycation of protein induces cancer cell proliferation and its inhibition by quercetin: spectroscopic, cytotoxicity and molecular docking studies. J. Biomol. Struct. Dyn. 39(3), 777–786 (2021). [DOI] [PubMed] [Google Scholar]
- 33.Niazvand F, Orazizadeh M, Khorsandi L, Abbaspour M, Mansouri E, Khodadadi A. Effects of quercetin-loaded nanoparticles on MCF-7 human breast cancer cells. Medicina (Kaunas) 55(4), 114 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Patel G, Thakur NS, Kushwah V et al. Mycophenolate co-administration with quercetin via lipid-polymer hybrid nanoparticles for enhanced breast cancer management. Nanomedicine 24, 102147 (2019). [DOI] [PubMed] [Google Scholar]
- 35.Yamada N, Matsushima-Nishiwaki R, Kozawa O. Quercetin suppresses the migration of hepatocellular carcinoma cells stimulated by hepatocyte growth factor or transforming growth factor-alpha: attenuation of AKT signaling pathway. Arch. Biochem. Biophys. 682, 108296 (2020). [DOI] [PubMed] [Google Scholar]
- 36.Carrasco-Torres G, Baltierrez-Hoyos R, Andrade-Jorge E, Villa-Trevino S, Trujillo-Ferrara JG, Vasquez-Garzon VR. Cytotoxicity, oxidative stress, cell cycle arrest, and mitochondrial apoptosis after combined treatment of hepatocarcinoma cells with maleic anhydride derivatives and quercetin. Oxid. Med. Cell. Longev. 2017, 2734976 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mizushima N. Autophagy: process and function. Gene Dev. 21(22), 2861–2873 (2007). [DOI] [PubMed] [Google Scholar]
- 38.Yun CW, Lee SH. The roles of autophagy in cancer. Int. J. Mol. Sci. 19(11), 3466 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy. Nat. Rev. Clin. Oncol. 8(9), 528–539 (2011). [DOI] [PubMed] [Google Scholar]; •


