I. Introduction
Cancer remains the second leading cause of disability and death worldwide where around 1 in 6 deaths are related to cancer. In 2020, around 19 million new cancer cases and almost 10 million cancer-related deaths were recorded globally [
1].
Lung cancer is a major public health concern, with significant impact on individuals and communities worldwide. Lung cancer has a high incidence rate; it was the second most commonly diagnosed cancer in 2020. Lung cancer is the leading cause of cancer-related deaths (around 1.8 million deaths or 18% of global cancer-related deaths in 2020) [
1]. With a 21% 5-year survival rate, lung cancer has one of the most poor prognoses [
2].
Depending on its progression, or the presence of metastasis, treatment of lung cancer will differ. Surgical resection, combination chemotherapy, and stereotactic body radiation are considered as the main modes of treatment. A significant obstacle in lung cancer therapy is the resistance that develops to chemo- and radio-therapy. Following therapy, resistance can lead to the relapse of lung cancer around 6 months post-therapy, in many patients [
3,
4]. Cisplatin is a cytotoxic chemotherapeutic agent used to treat several cancers including lung cancer [
5]. The standard of treatment for limited-stage lung cancer remains chemotherapy using four to six cycles of cisplatin and etoposide and concurrent radiation therapy. However, 60-70% of patients are at extensive-stage lung cancer and require a different treatment regimen; also involving cisplatin [
4,
6]. Nevertheless, these treatments can become refractory in patients. When relapse takes place lung cancer is usually refractory to treatment and has poor prognosis due to the limited availability of therapeutic options [
3,
4]. As a result, there is an urgent demand for new treatment options and alternative therapeutic modalities that can treat lung cancer and circumvent its chemoresistance. Herbal medicine represents a promising alternative in this regard.
Herbal medicine and phytotherapy, medicinal treatment based on plants or herbs and their extracts, have played a central role in the development of several anti-cancer agents such as Paclitaxel, Camptothecin, and Vincristine [
7,
8,
9,
10,
11]. Herbal medicine can be utilized as an assistant therapeutic modality as well. It can enhance the response rate to chemotherapy as well as radiotherapy, help overcome resistance to therapy, decrease the severity of side-effects caused by chemotherapy and radiotherapy, and enhance the quality of life and survival of cancer patients [
12,
13,
14,
15]. Relatedly, herbal preparations have been used as well to reduce the severe side effects of chemotherapy, including cancer-related fatigue [
7]. Herbal preparations and herbal active compounds act as anti-cancerous and anti-metastatic agents through several mechanisms, including scavenging of reactive oxygen species (ROS), modulation of epithelial to mesenchymal transition (EMT), impairment of angiogenesis, modification of the expression and activity of matrix metalloproteinases (MMPs), among others [
16,
17].
The Fabaceae family of plants includes a large number of domesticated species which are harvested as crops for human and animal consumption as well as oils, fuel, fertilizers, and medicinal and agricultural varieties [
18]. Medicago is a genus of the Fabaceae family and it comprises more than 83-87 different species of flowering plants [
18,
19,
20]. Twenty of these species are herbaceous perennials, 63 are herbaceous annuals and only 2 are shrubs [
20].
Medicago orbicularis L. Bartal. (common names: black disk, button medick, or button clover) is a species of the Medicago genus indigenous to Eurasia and North Africa [
19]. The plant is distributed throughout the Mediterranean basin mainly in Palestine, Lebanon, Syria, Algeria, Spain, France, Italy and Greece, in Middle East countries such as Iraq and Iran, and several other countries [
18].
M. orbicularis is a winter annual plant which flowers in Spring and early Summer. Its stems are leafy, stipules are fimbriate, leaflets are oval non-hairy with toothed margins, flowers are orange-yellow, seed pods are flat, coiled and lack spines [
19].
M. orbicularis phytochemical composition has not been investigated. But, other Medicago species have been reported to be rich in phytochemicals, including flavonoids, carotenoids, saponins, phenolic acids, and phytoestrogens [
21,
22,
23]. These phytochemicals have been demonstrated to exhibit anticancer activities through several mechanisms including apoptosis, antiproliferative, immunomodulatory, and anti-inflammatory activities, and the capacity to modulate oxidative stress [
24,
25,
26,
27,
28]. It is well recognized that antioxidant activity is one mechanism through which medicinal plants may exert a therapeutic effect [
27,
28,
29,
30,
31,
32]. Close-by species such as
M. polymorpha, M. sativa, M. arabica, or M. truncatula were shown to possess a robust antioxidant activity [
22,
23]; an indication to the potential anti-oxidant and therapeutic value of
M. orbicularis. Güleç and Kültür reported a traditional therapeutic use of
M. orbicularis in Turkey for heart diseases [
33]. However, there are no studies currently on the antioxidant potential, therapeutic applications or anti-cancerous properties of
M. orbicularis. Therefore, we assessed the antioxidant capacity, hemolytic properties, phytochemical composition and the effects of
M. Orbicularis on cell proliferation, cell aggregation, and cell migratory potential against A549 human lung cancer cells. Additionally, the combinatorial effect of a treatment using both cisplatin and
M. orbicularis on A549 cells was tested.
II. Materials and Methods
Cell Culture and Reagents
Human lung adenocarcinoma cell line A549 and primary normal neonatal fibroblast (HDFn) were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). Cells were maintained in a humidified chamber at 37 °C and 5% CO2 in DMEM (Dulbecco’s Modified Eagle’s Medium; cat# D0819 Sigma-Aldrich Co., St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS) (cat# F9665, Sigma-Aldrich) and 100U/ml penicillin and 0.1 mg/ml streptomycin (cat# P4333, Sigma-Aldrich). Cells were passaged by trypsinization when they reached 90% confluence.
Plant Collection and Extraction
Fresh
Medicago orbicularis L. Bartal was collected from Almat, Byblos, Lebanon between May-June 2021. A voucher specimen was stored in the herbarium of the Faculty of Pharmacy, Lebanese University, Beirut, Lebanon and was authenticated by Professor George Tohme, an experienced taxonomist and herbalist.
Medicago orbicularis L. Bartal: Kingdom Plantae; Phylum Tracheophyta Class Magnoliopsida; Order Fabales; Family Fabaceae; Genus Medicago L. (Index Kewensis;
https://powo.science.kew.org/taxon/urn:lsid:ipni.org:names:506322-1).
Extracts were obtained from leaves, stems and fruits of M. orbicularis. In brief, the different plant parts were cleaned, air-dried in the shade at room temperature, ground to powder, and kept in plastic containers, away from light, heat and moisture. Then, 10 grams of the powder were mixed with 100 ml of 70% aqueous ethanol, and the mixture was placed in a reciprocating shaker and continuously agitated at 150rpm for 3 to 4 days. The solutions were filtered and concentrated by a rotary evaporator at 40°C and then lyophilized using a lyophilizer. The lyophilized extracts were stored at -20°C until their use. At the time of use, the extracts were dissolved in 70% ethanol.
DPPH (α, α-diphenyl-β-picrylhydrazyl) Antioxidant Activity
The antioxidant free radical scavenging ability of the ethanolic extracts of the different parts of
M. orbicularis (leaves, fruits and stems) was measured using the DPPH radical scavenging assay which was performed as described [
34]. DPPH changes color from intense purple to pale-yellow due to the donation of a hydrogen atom, if the plant extract has radical scavenging activity. For
M. orbicularis ethanolic extracts, 1 ml of different concentrations of the ethanolic plant extracts (50, 100, 150, 200, 300 or 500 μg/ml) was mixed with an equal volume of DPPH (cat# D9312, Sigma-Aldrich Co.) solution (0.15 mM in ethanol). The blank consisted of 1 ml of DPPH solution and 1 ml of 70% ethanol. Mixed samples were then kept in the dark for 30 min and the OD was measured at a wavelength of 515 nm using a spectrophotometer. Ascorbic acid, a potent antioxidant, was employed as a positive control and reference. The DPPH scavenging activity of each concentration of the extracts was calculated using the formula:
In vitro Cytotoxicity Assay
In vitro cytotoxicity of M. orbicularis extracts was assessed using MTT assay which is a reduction assay that measures cellular metabolic activity and is reflective of cell viability. A549 and neonatal fibroblast cells were seeded in a 96-well tissue culture plate at a density of 0.7x104 cells per well in DMEM culture medium containing 10% FBS and penicillin/ streptomycin. After an overnight incubation, cells were treated with different concentrations (50, 100, 150, 200, 300 or 500 μg/ml) of M. orbicularis extracts for 24, 48 and 72 h. As a control, cells were treated with a concentration of ethanol (vehicle-control cells) equal to the concentration of ethanol in the extract-treated cells. Following the treatment, MTT reagent (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyl Tetrazolium Bromide; cat# M5655, Sigma-Aldrich Co.) was added for 3 hours until a purple formazan precipitate was formed. Media were removed gently, and an isopropanol-HCl solvent was added to dissolve the formazan precipitates. The Optical Density (OD) was measured by an ELISA plate reader at a wavelength of 595 nm. The percentage of cell viability of the treated cells was calculated using the following formula:
The IC50 (half-maximal inhibitory concentration) value, was determined by extrapolation from the cell killing curve to determine the concentration of the extract that induced 50% cell death.
Cisplatin (Sigma-Aldrich Co.), dissolved in DMSO, was used as a positive control of the cytotoxic effect of M. orbicularis parts extracts on A549 cells. A549 cells were treated for 24, 48 and 72 hours with increasing concentrations of cisplatin (2.5, 5, 10 and 20 μg/ml) and compared to the DMSO treated vehicle-control cells. IC50 values of cisplatin in A549 cells were determined at the three time points. Data are displayed as the mean ± SEM of three independent experiments.
Cell Aggregation Assay
Seeded A549 cells were detached using 2mM EDTA PBS (calcium- and magnesium-free). Cells were pelleted and washed with PBS, and then resuspended in 1ml PBS including or not 100 or 150 μg/ml of M. orbicularis fruit extract alone, 5 μg/ml cisplatin alone, or a combination of both and kept shaking on a rocker for 60 minutes at 37°C in a cell culture incubator. Cells were fixed with 1ml of 1% formaldehyde and imaged using an inverted microscope (Leica Microsystems GmbH, Wetzlar, Germany). Percentage of aggregation was calculated using the formula: ; where Nt is the number of single cells in treated wells and Nc is number of single cells in the vehicle control cells.
Assay of Hemolysis and Anti-hemolytic activity
Hemolytic activity assay was performed as previously described [
35]. Erythrocytes were obtained after separation from plasma by centrifugation at 2500 rpm for 10 min at 4°C starting from fresh sheep blood. Briefly, RBCs were washed three times with 1X PBS then pelleted and resuspended in PBS to obtain a 5% suspension of RBCs. Then, 50 μL of different concentrations (10, 20, 40, 50 and 100 μg/ml) of ethanolic extract of fruits of
M. orbicularis were added to 1 ml of the RBC suspension. The mixture was incubated at 37°C for 1.5 hours, then the suspension was centrifuged at 2,500 rpm for 10 min at 4°C and the OD measured at 540 nm using a spectrophotometer. Ethanol concentrations equal to those present in the fruit extracts were added to blood and used as a negative control, 1% SDS as a positive control of hemolysis of RBCs, and 1X PBS as the blank. Hemolytic levels were calculated as follows:
Anti-hemolytic activity assessment was performed as previously published [
36]. Briefly, the same steps were performed as in the hemolysis assay except that H
2O
2 was added to induce hemolysis. After adding
M. orbicularis extracts to the RBC suspension for 20 min, RBCs were incubated with 350 μL of 30% H
2O
2 at 37°C for 1.5 h. The suspension was centrifuged at 2500 rpm for 10 min at 4°C and OD at 540 nm was determined. Thirty % H
2O
2 was used as a positive control, and 1X PBS as the blank. Anti-hemolytic levels were calculated according to the following equation:
Scratch/ Wound-Healing Assay
A549 cells were grown in 12-well tissue culture plates until confluence. A scrape was made through the confluent monolayer using a sterile yellow 200 μL pipette tip. The culture medium was then removed, the cells were washed twice with PBS (Sigma-Aldrich, St. Louis, MO, USA) to remove cellular debris, and incubated at 37℃ in fresh medium in the presence or absence of the indicated concentrations of M. orbicularis extracts. Photomicrographs of the scratch were obtained at baseline (0 h) and for the next 4 to 12 h using an inverted microscope (objective 10×). The width of the wound was expressed as the average ±SEM of the difference between the measurements taken at time zero and the corresponding time points. Ethanol was used as a vehicle control.
Western Blotting Analysis
A549 cells were seeded in a 6-well plate at a density of 2.8x105 cells per well and cultured for 24 h. The cells were treated with extracts of M. orbicularis for 48 h and then the cells were washed twice with PBS and lysed using a lysis buffer containing 2% SDS, 60 mM Tris lysis buffer (pH 6.8), and protease inhibitors and centrifuged at 10,000 g for 10 min. The protein concentration of the supernatants was determined using the Bradford protein assay kit (Biorad, Hercules, CA, USA) and 25 μg of protein lysates were resolved on 10% SDS-PAGE before being transferred to a polyvinylidene difluoride membrane (Immobilon PVDF; Biorad, Hercules, CA, USA). The membranes were then blocked for 1 h at room temperature with 5% non-fat dry milk in TBST (TBS and 0.05% Tween 20). Immunodetection was performed by incubating the membrane overnight with specific primary antibodies at 4℃. Primary antibodies included rabbit IgG anti-full length human poly-adenosine diphosphate (ADP) ribose polymerase-1 (PARP-1) (Cell Signaling Technologies, CST 9542; dilution 1/1,000), rabbit anti-human proCaspase 3 (Santa Cruz, sc-7148; dilution 1/1,000), rabbit anti-human B-cell lymphoma 2 (BCL-2) (Abcam, ab32124; dilution 1/5000), rabbit anti-Bcl-2 associated X protein (BAX) (Abcam, ab32503; dilution 1/2,000), and rabbit anti-GAPDH (Abcam, ab181602; dilution 1/10,000). Membranes were washed and incubated with a horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG secondary antibody (Abcam, ab6721; dilution 1/5,000) for 1 h followed by washing in TBST. Immunoreactive bands were detected using ECL substrate kit (Thermo Scientific, Rockford, IL, USA), and membranes were scanned using the Chemidoc imaging system (Bio-Rad). The intensity of the obtained bands was quantified using ImageJ software (NIH, MD, USA). All bands were normalized to GAPDH, which was used as a loading control.
Spectroscopic determination of total phenolic content
Total phenolic compounds content (TPC) was determined using the Folin–Ciocalteu colorimetric oxidation/reduction-based reaction as previously described [
37], with slight modifications. Briefly, 100µl of diluted fruit extracts of
M. orbicularis were added to 500µl of Folin–Ciocalteu reagent, followed by the addition of 400µl of saturated sodium carbonate solution (2%). The oxidation products of the reaction show a blue color with a broad light with an absorption maximum of 765 nm. OD 765 of the different mixtures was measured, against a blank, using a spectrophotometer. A calibration curve was prepared using a strong antioxidant, gallic acid, and the results were expressed as gallic acid equivalents (GAE) in milligrams per gram of the extract.
Spectroscopic determination of total flavonoids content
The total flavonoids content (TFC) of
M. orbicularis fruit extracts was determined by the aluminium chloride colorimetric method as previously detailed [
38]. AlCl
3 forms a flavonoid–aluminium, complex that has a maximum OD at 510 nm. Briefly, a 100µl aliquot of appropriately diluted fruit extracts of
M. orbicularis were added to 400µl of distilled water. Then, 400µl of 5% NaNO
2 were added, followed by 300µl of 10% AlCl
3. After 6 min of incubation 200µl of 1M NaOH were added to the mixture followed by 240µl of distilled water. The absorbance of the resulting pink solution was determined at 510 nm versus a blank (water). A calibration curve was prepared using a known flavonoid, quercetin. The curve was used to calculate the amount of flavonoids in the plant extracts which was expressed as quercetin equivalents in mg/g dry weight of the plant part.
Gas Chromatography/ Mass Spectrometry (GC/MS)
The phytochemical compounds of M. Orbicularis fruit extracts were analyzed using GC/MS analysis. A Perkin Elmer Clarus 680 (Perkin Elmer, USA) system attached to a triple quadrupole mass spectrometer was used. Chromatography was conducted on a hydrophobic capillary column RTxi-5 Sil MS column (30 m × 0.25 mm ID × 0.25 µm) using an injection volume of 10 µl, a flow rate of 1.5 mL/min, a pressure of 23.1 KPa, and an average velocity of 0.2 sec. The temperatures of the source and the interface were 200 °C and 280 °C, respectively. The initial temperature was set at 80 °C for 2 min, increased to 250 °C at 15 °C/min, and raised to 280 °C at 15 °C/min (held for 12 min). Identification of phytochemicals in extracts was carried out by comparing the obtained retention indices with those of chemical compounds in the database of the National Institute of Standards and Technology (NIST).
Statistical Analysis
Results were evaluated for statistical difference using One-way ANOVA followed by Bonferroni test to calculate P values. Data are presented as mean ± standard error of the mean (SEM) and a p-value of P <0.05 was considered as statistically significant. Statistical analysis was performed using GraphPad Prism 8 software (GraphPad Software Inc., San Diego, CA, USA).
IV. Discussion
Cancer is a complex and devastating disease which affects millions of lives worldwide. Lung cancer, particularly non-small cell lung cancer, has the highest mortality rate among malignant tumors across the globe [
43]. Current treatments for lung cancer such as chemotherapy and radiotherapy are associated with considerable toxicity and other side effects. In addition, lung cancer patients develop resistance to therapy and suffer from lung cancer recurrence as early as 6 months following therapy [
3,
4]. In this regard, there is a revived research interest in using herbal- and plant-based therapies as sources of anti-cancer bioactives. Several herbal remedies have been shown to have efficacy and minimal toxicity and side effects for disease treatment [
12,
13]. In addition, plants and their phytochemicals are being used as starting scaffolds for developing more potent anticancer agents [
15,
30]. Historically, plants or herbs and their extracts were used for the development of several important anti-cancer agents such as Paclitaxel, Camptothecin, and Vincristine [
7,
8,
9,
10,
11,
30]. Furthermore, several intricate biochemical pathways are simultaneously dysregulated in cancer [
6,
15], and herbal-based or herbal-derived remedies can target several molecular pathways and can become an alternative or complementary treatment to conventional cancer therapies [
7,
12,
13,
14,
15,
53,
54]. Relatedly, multiple recent
in vitro and
in vivo studies demonstrated that concurrent use of natural products with conventional treatments (chemo- and radio-therapy) can synergistically sensitize tumors to therapy, enhancing therapeutic efficacy and reducing toxicities [
15]. Many studies have documented the promising use of herbal plant extracts against cancer cell lines
in vitro and
in vivo in animal models of cancer, and are now being tested in clinical trials. These studies have shown that herbal-based remedies have antioxidant, antihemolytic, and apoptosis-inducing effects. They can also impact cell proliferation, aggregation and adhesion, migration, and metastasis [
16,
17,
55].
Medicago orbicularis L. Bartal is an understudied plant species as far as its therapeutic effects are concerned. Except for a study reporting a traditional use of
M. orbicularis for heart diseases management in Turkey [
33], there are no reports on the therapeutic applications or phytochemical composition of
M. orbicularis. Notably, M. orbicularis anti-cancerous activities have not been explored yet [
18]. It was therefore pertinent to study phytochemical composition, antioxidant capacity, antihemolytic properties, and the effects of
M. orbicularis on cell proliferation and apoptosis, cell aggregation, and cell migratory potential against human lung adenocarcinoma A549 cells, in addition to the combinatorial effect of a treatment of both the chemotherapeutic agent cisplatin and
M. orbicularis on A549 cells.
There are no studies currently on the anti-oxidant potential of
M. orbicularis, but other species of the genus Medicago such as
M. polymorpha, M. sativa, M. arabica, and M. truncatula were shown to have potent anti-oxidant activities and to be rich in phytochemicals [
22,
23]. Since anti-oxidant activity is often times accompanied by therapeutic effects [
27,
28,
29,
30,
31,
32], this study was initiated with an evaluation of the antioxidant activity of the ethanolic extract of different plant parts of
M. orbicularis including its leaves, fruits and stems. All plant parts of
M. orbicularis showed anti-oxidant potential and the leaves showed the highest antioxidant activity.
To test if the anti-oxidant capacity of
M. orbicularis is accompanied by an anti-cancerous effect, we tested the cytotoxic effect of
M. orbicularis (leaves, fruits and stems) on A549 human lung adenocarcinoma cells. All
M. orbicularis plant parts inhibited proliferation of A549 cells, but
M. orbicularis fruits exhibited the highest reduction of viability of A549 cells and were chosen to perform the rest of the experiments of the study. The National Cancer Institute (NCI, USA) considers an IC50 of 30 μg/mL to indicate a strong cytotoxic activity and a promising candidate for the further purification of a crude extract, and an IC50 of 31–200 μg/ml to indicated moderate cytotoxicity [
56]. The IC50 of the fruit extracts at 72 h of treatment was 86.18± 1.93 μg/mL, indicating that this crude extract has moderate cytotoxicity. To date, no other study has reported the cytotoxicity levels of
M. orbicularis against cancerous cells. Importantly, the cytotoxic effects of the fruit extracts were showed selectivity to A549 cancerous cells and did not affect human neonatal fibroblast cells even at high concentrations. This indicated that
M. orbicularis fruits extracts may not have side effects when used
in vivo.
Plant bioactives can decrease viability of cancer cells through several mechanism including induction of apoptosis [
7,
12,
13,
14,
15,
53,
54]. Cytotoxic agents can cause an increase in the expression of the pro-apoptotic protein BAX that makes pores in the mitochondrial membrane leading to the release of cytochrome C [
57]. Release of cytochrome C initiates the execution phase of apoptosis and activates Caspase 9 which cleave pro-Caspase 3 into active Caspase 3, the main executer effector caspase [
58,
59]. Active Caspase 3 can cleave many protein substrates [
60], such as caspase activated DNAse that fragments genomic DNA [
61] and PARP-1, to promote apoptosis [
59]. PARP-1 is required for DNA repair where it adds poly (ADP ribose) polymers to acceptor proteins to promote repair of DNA damage. PARP-1 cleavage by Caspase 3 inactivates PARP-1 leading to reduction of DNA repair and promotion of apoptosis [
58,
59,
62,
63]. These events are recognized as the intrinsic apoptotic pathway. The anti-apoptotic protein BCL-2 can inhibit the release of cytochrome C into the cytoplasm, thereby attenuating the intrinsic apoptotic pathway [
59,
64]. Western blotting analysis of lysates of A549 cells treated with 100 and 150 g/ml
M. orbicularis fruit extract indicated that M. orbicularis fruit extracts activated Caspase-3, inactivated PARP-1, and lowered the expression of BCL-2 and increased the expression of BAX, suggesting the activation of the intrinsic apoptotic pathway. The results showed a significant decrease in the protein levels of full-length PARP-1 and proCaspase 3 in A549 cells in a dose-dependent manner. Moreover, there was a dose dependent decrease in the BCL-2/BAX ratio. This is the first report showing that
M. orbicularis-induced death of A549 cells is mediated, at least partly, by the intrinsic apoptotic machinery. Other mechanisms of cell death, such as autophagy, necroptosis, or pyroptosis, may also be induced by
M. orbicularis, and need to be tested in future studies.
As part of the safety profile of a cancer drug, its systemic infusion through the blood of a patient should not cause any hemolysis of red blood cells [
65].
M. orbicularis fruit extracts did not cause any hemolysis of RBCs. Even more, the fruits extracts protected RBCs against hemolysis. This attested to the possible safety of these extracts in future
in vivo or clinical studies.
Metastasis is the major culprit behind cancer-associated mortality [
66,
67,
68]. Cell migration is a key requirement for the metastatic process. Although cell migration is essential in many physiological processes such as wound repair, tissue formation, and proper immune response, its deregulation is required for the initial steps of cancer metastasis as cells spread away from the primary tumor site. Cell migration involves the downregulation of cell junction and adhesion proteins and proteolysis of ECM proteins by metalloproteinases (MMPs) [
67,
69] . We assessed the effect of the fruit extract on migration of A549 using a wound healing assay. Non-cytotoxic concentrations of the fruit extracts of
M. orbicularis inhibited migration of A549 cells in a concentration-dependent manner. This indicated that M. orbicularis fruits could attenuate the malignant phenotype of A549 cells by inhibiting cell migration and consequently metastasis. This result invites future investigation of the ability of
M. orbicularis to inhibit the invasive potential of A549 cells through its action on MMPs, and proteins of the process of epithelial-to-mesenchymal transition (EMT), a hall mark of carcinogenesis and metastasis [
27,
28,
67,
68,
70].
In order for cancer cells to migrate and metastasize, they should lose their adhesion to neighboring cells allowing them to migrate and invade at secondary tumor sites and organs [
27,
28,
67,
68,
70]. Cancer therapeutics can act by strengthening cell adhesion and aggregation to prevent cell migration and metastasis [
71]. In this study, an aggregation assay showed that
M. orbicularis fruits significantly enhanced cell aggregation, signifying that the extract enhanced adhesion of A549 cells. This result further attests to the ability of M. orbicularis to attenuate the malignant phenotype of A549 cells. Future, studies should focus on elucidating the molecular mechanisms underlying this finding, include examination of cell adhesion proteins, connexins gap junction proteins, and EMT markers such as E-cadherin, N-cadherin, vimentin, Snail, among others [
72].
Apart from their direct suppressive effects on carcinogenesis and cancer metastasis, herbal remedies and plant extracts have been used to complement conventional therapies since they can target several molecular pathways other than those targeted by conventional therapy, and therefore may overcome refractoriness that develops against cancer therapeutics [
7,
12,
13,
14,
15,
53,
54]. When combined with chemotherapy, plant-based therapies can enhance response to chemotherapy, help overcome resistance to therapy, decrease the severity of side-effects of chemotherapy including cancer-related fatigue [
7,
12,
13,
14,
15]. Chemotherapeutic agents, including cisplatin can become refractory in lung cancer patients, and as result the cancer may relapse with poor prognosis [
3,
4]. Combination of cisplatin with a multitargeted herbal remedy may circumvent this problem. In this study, the cytotoxic effect of cisplatin alone on A549 cells was confirmed. The IC50 of cisplatin with A549 that we obtained are similar to those reported in the literature [
43]. In order to determine the impact of a combination treatment on cisplatin-induced death of A549 cells, we applied a co-treatment of cisplatin and the ethanolic extract of
M. orbicularis fruits. The results showed that the combination treatment significantly augmented cisplatin-induced decrease in cell viability of A549 cells, when compared to either the extract alone, or cisplatin alone. These results show that
M. orbicularis may be a source of a complementary therapy for lung cancer, in combination with chemotherapy. The combination treatment showed a similar result when tested on aggregation of A549 cells. Co-treatment of
M. orbicularis fruits extracts and cisplatin showed that the combination treatment significantly augmented the increase in aggregation of A549 cells, when compared to either the extract alone, or cisplatin alone. Taken together, these results support
M. orbicularis as a source for development of anti-lung cancer drug candidates that can complement chemotherapy.
M. orbicularis phytochemical composition has not been defined yet. But, other Medicago species have been reported to be rich in phytochemicals [
21,
22,
23], which have anticancer activities [
24,
25,
26,
27,
28]. Natural polyphenols are plant secondary metabolites which have two or more phenol rings. Polyphenols health benefits include being antioxidant, antidiabetic, cardioprotective, neuroprotective. Relatedly, phenolics have been reported to have strong anticancer effects through various mechanisms including removal of cancer cells, inhibition of cell cycle, and induction of apoptosis, inhibition of metastasis among others [
52]. Flavonoids are natural polyphenols with documented anticancer and antioxidant properties. Flavonoids have been reported to inhibit carcinogenesis by suppressing oxidative stress through their antioxidant activities [
26,
51]. Our results indicated that
M. orbicularis fruits ethanolic extracts have strong antioxidant and cytotoxic effects against A549 cells. These effects could be mediated by polyphenols and flavonoids of
M. orbicularis fruits extracts among other phytochemicals. In this study,
Medicago orbicularis fruits were shown to be rich in phenolics and flavonoids, which may explain the anti-oxidant and anticancer properties of
Medicago orbicularis fruits against A549 cells.
To further evaluate the phytochemical composition and establish a more comprehensive profile of the phytochemical constituents of
Medicago orbicularis fruits, GC-MS analysis was conducted. We identified 18 prominent peaks that correspond to bioactive compounds of
Medicago orbicularis fruits.
Medicago orbicularis ethanol extract was found to be a complex mixture of various classes of phytochemicals, including flavonoids, alkaloids, diterpenes, triterpenes, sesquiterpenes, sterols, alcohols, aldehydes, fatty acids, and hydrocarbons. Among the diverse array of phytochemicals present in the extract, several compounds were correlated with therapeutic properties, including anti-cancerous activities. For example, lupeol and its derivatives, which were the most frequently identified compounds, have been recently demonstrated to possess a diverse array of pharmacological activities [
73]. These activities included anticancer, antimicrobial, and antidiabetic effects, with certain lupeol derivatives exhibiting greater potency than lupeol [
74]. Future investigation should focus on more comprehensive assessment of the plant extracts of
M. orbicularis, using more advanced techniques such as preparative HPLC followed by LC-MS/MS or NMR, to identify the bioactives responsible for the therapeutic effects of crude extracts from this plant.
In conclusion, this study shows for the first time that the hydroalcoholic extract of M. orbicularis has anti-oxidant, anti-hemolytic, and cytotoxic properties against A549 lung cancer cells. The decrease in cell viability could be at least partly mediated by the intrinsic pathway of apoptosis, involving PARP-1, Caspase 3, and BCL-2/BAX. M. orbicularis was also able to suppress cell migration and enhance cell aggregation of A549 cells. These activities could be attributed to the phytochemicals present in M. orbicularis such as polyphenols and falvonoids, and the other compounds that were identified by GC/MS. These properties place M. orbicularis as potential source of new candidate effective natural agents for the treatment of the lung cancer.