1. Introduction
Oral cancer is one of the public malignancies worldwide that accounts for 2–4% of all cancer cases, with nearly one million new cases diagnosed annually. Indeed, oral cancer ranks eighth among the most common causes of cancer-related deaths. Oral squamous cell carcinoma (SCC) is an aggressive global malignancy constituting more than 90% of diagnosed oral cancer cases [
1]. Most cases occur in males over the age of 50, but the incidence in women and young patients is listed to be increasing [
2]. Oral SCC is frequently detected in late advanced stages and has a poor prognosis with a 50–60% five-year survival rate [
3]. Risk factors for oral SCC include age, gender, alcohol, tobacco, diet, and nutrition, with tobacco smoking being the most common risk factor [
4].
Treatment strategies for oral SCC have significantly enhanced in the past few years; however, the survival rate is still poor, especially in advanced cases. Chemotherapy, radiotherapy, and surgery are currently conventional therapies for oral SCC. Treatment options vary mainly depending on the disease stage. Chemotherapy is usually advised for patient’s ineligible for surgery due to extensive local tumor growth, medical comorbidities, or distant metastasis. Chemotherapy has been used alone as a primary treatment modality or as a radiation sensitizer in combination with radiotherapy. Of note, combination therapy has been reported to increases the five-year total survival rate by 6.5% compared with a single modality treatment [
5]. Indeed, combination therapy has been shown to be more effective in advanced oral SCC [
6]. However, this efficacy was limited by high rate of recurrence, chemoresistance to some anticancer drugs, and harmful side effects [
7].
Among current chemotherapies, cisplatin or cis-diamminedichloroplatinum (II) (CDDP) is a first-generation anticancer agent that has been commonly used for several years to treat oral SCC. Cisplatin triggers cytotoxicity through binding to DNA, the creation of DNA adducts, and interfering with the cell repair leading ultimately to apoptosis. However, the rapid development of both inherent and acquired chemoresistance to cisplatin restricts its clinical application [
8]. So, recent studies are directed to find new therapeutic strategies, including combination drug regimens for oral SCC treatment [
9].
The condensation reaction of primary amine and carbonyl compounds makes Schiff bases. Schiff bases have a wide range of biological activities comprising anti-inflammatory, antimicrobial, and anticancer activities [
10]. Their DNA-binding and DNA cleavage activities made-up the use of these compounds as potential drugs in cancer treatment. The Schiff bases antitumor activity has been related to its ability to intercalate DNA and interfering with the role of the topoisomerases, that regulate the DNA topology during the cell division [
11]. Schiff bases possess the ability to stabilize various metal ions in different oxidation states, forming complexes. Multiple studies have demonstrated mild to good cytotoxicity of Schiff bases against various malignant tumors. Metal inclusion in the complex has a significant impact on the anticancer effectiveness of the Schiff bases [
12]. Specifically, Schiff base heterodinuclear Cu(II)–Mn(II) complex is a novel Schiff base metal derivative with reported therapeutic efficacy against liver and colon cancer [
13].
Tumor-derived cell lines as model systems with resemblances to the original endogenous human tumors are suitable resources currently used as screening platforms for estimating the potential efficacy of anticancer therapeutics and understanding the underlying biological processes in carcinogenesis [
14]. Consequently, cell lines derived from multiple tumor tissues are currently aiding in identification of the events linked to the development of cancer to help the generation and establishment of therapies alternative to the current defective modalities [
15].
Stem cell research has become an advantageous and advanced scientific research field. Stem cell research pushes far beyond the therapeutic and scientific potential of regenerative medicine. Currently, the field of stem cell research is attracting enormous attention for application in drug screening [
16]. Dental stem cells are a good source of adult stem cells that are merely available by tooth extraction or during the replacement of primary tooth. Oral-derived gingival mesenchymal stem cells (GMSCs) are self-renewing and easily accessible mesenchymal adult stem cells widely applied in oral and maxillofacial diseases [
17].
The present study was designed to assess the anticancer activity of Cu(II)–Mn(II) Schiff base tetradentate complex (see
Supplementary Material) alone and in combination with cisplatin against oral squamous cell carcinoma (SCC) cell line using GMSCs as control. This was performed through in vitro assessment of cytotoxicity, induced apoptosis and gene and protein expression of apoptotic proteins.
3. Materials and Methods
All experimental procedures were performed according to protocols of the Ethics Committee of the Faculty of Dentistry, Mansoura University, and the animal house unit of Nile Center for Experimental Research (NCER), Mansoura city, Egypt.
3.1. Schiff Base Cu(II)–Mn(II) Complex Preparation
Manufacture and characterization of heterodinuclear Cu(II)–Mn(II) Schiff base tetradentate complex was performed according to the process described by Dede et al. [
18]. The prepared compound was then preserved in a light protected tube at room temperature until use. A stock solution of the Schiff base complex was prepared by dissolving 1 mg per 1 mL dimethyl sulfoxide (DMSO, Sigma-Aldrich, St. Louis, MO, USA). Then various concentrations were prepared by dilution in culture media RPMI-1640 (RPMI, Invitrogen, Gibco, Paisley, UK) and Dulbecco’s modified Eagle’s medium (DMEM, Invitrogen, Gibco, Paisley, UK) supplemented with 1% penicillin/streptomycin/amphotericin (PSA, Invitrogen, Gibco, Paisley, UK) and 10% fetal bovine serum (FBS, Invitrogen, Gibco, UK). 10% (
w/
v) stock solution of cisplatin (cis-Diammine-di-chloroplatinic (II), cis-DDP), (Medac, Wedel, Germany) was prepared in phosphate-buffered saline (PBS, Pan-Biotech, Aidenbach, Germany). Then, 1% in DMEM was used for dilution. For combination treatment, 250 µM Schiff base complex was combined with 1.0 µg/mL cisplatin.
3.2. Isolation and Culture of Gingival Margin-Derived Stem Cells (GMSCs)
All in vitro steps were performed at Nile Center for Experimental Research (NCER), Mansoura city, Egypt (Stem Cell Unit). Isolation of gingival stem cells was according to a protocol described by Zhang et al. (2009) [
19]. Four pathogen-free albino rats weighing 80–150 g were euthanized and rat tissue samples were collected from clinically healthy gingiva. To separate the epithelial and lower spinous layer, the samples were treated aseptically and incubated with dispase (2 mg/mL; Sigma-Aldrich) overnight at 4 °C. Thereafter, the connective tissues were minced into 1–3 mm
2 fragments and digested in sterile phosphate-buffered saline (PBS) containing 4 mg/mL collagenase IV (Worthington Biochemical) for 2 h at 37 °C. The samples were then grown in Dulbecco’s Modified Eagle’s medium Ham’s F-12 (DMEM-F 12) with 1% antibiotic (streptomycin, penicillin, and amphotericin) and 10% fetal bovine serum (FBS). The explants were maintained in the incubator (5% CO
2 humidified air and 37 °C) for 14–21 days to reach 80% confluency. The cells were then trypsinized and subcultured (
Figure 8a).
3.3. Oral Squamous Cell Carcinoma Cell Lines (SCC) and Culture
H357 (Human oral squamous cell carcinoma, tongue) cell lines were obtained from Sigma-Aldrich Co. (St. Louis, MO, USA). Eagle’s Dulbecco’s modified medium was used as a cell culture medium enhanced with 100 IU/mL penicillin, 10% fetal bovine serum, and 100 μg/mL streptomycin. The cells were cultured at 37 °C under a humidified atmosphere containing 5% CO
2. Periodic examination of cultured cells was performed by examining the cultured cells at a 400× magnification under a light microscope without preliminary fixation using an Olympus microscope (New York Microscope Company, Hicksville, NY, USA),
Figure 8b.
3.4. Cell Characterization of GMSCs
Flow cytometry was used for determination of the immunophenotype. Gingival cells (5 × 105) were incubated at room temperature for 20 min in the dark with individual fluorescein isothiocyanate (FITC)-conjugated primary monoclonal antibodies in 100 μL PBS. CD45, CD90, CD105, and CD34 were used as primary antibodies (Abcam, Cambridge, UK). Cells were then diluted in 2 mL PBS/bovine serum albumin followed by centrifugation and resuspension in 200 μL of paraformaldehyde 4%. BD Accuri C6 flow cytometer and BD Accuri C6 software programs were used for acquisition and analysis (BD Biosciences, San Jose, CA, USA).
3.5. Study Groups
Cell culture samples were divided into the following four groups: Group I non-treated control, which was subdivided into Ia (control GMSCs) and Ib (control SCC). Group II Schiff base complex (SB) treated, which was subdivided into IIa (GMSCs with SB) and IIb (SCC with SB). Group III cisplatin (cis-DDP) treated, which was subdivided into IIIa (GMSCs with cis-DDP) and IIIb (SCC with cis-DDP). Group IV treated with both Schiff base complex and cisplatin, which was subdivided into IVa (GMSCs with both SB and cis-DDP) and IVb (SCC with both SB and cis-DDP). The cells were treated with different concentrations of cisplatin and Schiff base complex, and then the cytotoxicity was observed by MTT assay. IC50 values were calculated, and the effective dose was used for this study.
3.6. Cell Viability and Proliferation Analysis
Reduction of tetrazolium dye MTT [3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide] [
20] by viable cells was used to assess proliferation rate in response to treatment (MTT cell growth assay kit, Funakoshi, Japan). Cells were seeded in 96-well plate (Falcon; Lincoln Park, NJ, USA) at a density of 2 × 10
3 in culture media (DMEM containing 10% FBS). The plate was incubated for 48 h, then various concentrations of cisplatin (0.5, 1.0, 1.5, 2.0 and 2.5 µg/mL) and Schiff base complex (150, 300, 450, 600 and 750 µM) were added and kept for 48 h. Following treatment, 10 μL of MTT solution was added to each well, and the plate was left for a further 4 h [
21]. The media was aspirated and acid isopropyl alcohol was added to each well to solubilize formazan crystals and the developed color was recorded at 550 nm was performed using microplate Reader (MTP300, Tokyo, Japan). Six cultured wells were used for each drug concentration, and the experiment was replicated 3 times. The 50% inhibitory concentration (IC50) was calculated using Graph Pad Prism Programming.
3.7. Annexin- V-FITC Binding Assay
Apoptotic cells were stained and counted by flow cytometry using Annexin V-FITC apoptosis detection kit (Sigma-Aldrich, St. Louis, MO, USA). Following treatment, cells from control and treated wells were washed with PBS two times and suspended at a concentration of 1 × 10
6 cells/mL in a one-fold binding buffer. Thereafter, 500 μL of each cell suspension was mixed with 5 μL of Annexin V FITC Conjugate and 10 μL of propidium iodide solution. The tubes were then incubated for 10 min at room temperature, and the reaction was assessed by flow cytometry instrument (FACStar caliber, Becton Dickinson, San Jose, CA, USA) using a filter over 600 nm for PI detection and 488 nm excitation and 515 nm bandpass filter for FITC detection. Annexin-v-positive/PI-negative cells were considered as early apoptotic cells, whereas Annexin-v-positive/PI-positive cells were reflected as late apoptotic cells, and the percentage of each phase was calculated with the software Cell-Quest [
22].
3.8. Apoptotic Makers (Caspase-3, Bcl-2 and Bax) Assay
Cellular detection of the apoptotic markers caspase-3, bcl-2 and Bax activity was determined by FCM employing the fluorescein isothiocyanate conjugated monoclonal antibodies (caspase 3 FITC, Rabbit anti- active caspase 3 cat. No, 559341), (PE Hamster Anti-Mouse Bcl-2 Set Cat No. 556537) and (Bax Monoclonal Antibody (6A7) Cat No. MA5-14003) according to the manufacturer instructions. Following treatment, cells from control and treated wells were washed with 2 mL PBS/BSA (Bovine serum albumin) then centrifuged at 2000 rpm for 5 min. The supernatant was discarded and the pellet was re-suspended in 100 µL of PBS. Ten µmicroliters of the 1st antibody were mixed well then incubated for 30 min at room temperature in dark. Cells were then washed with 2 mL PBS/BSA, centrifuged at 2000 rpm for 5 min and supernatant was discarded. Ten µmicroliters of conjugated IgG secondary antibody was added and then the tubes were incubated for 20 min. at room temperature in dark. Finally, cells were re-suspended in 200 µL of 4% par formaldehyde in PBS. The percentage of each apoptotic marker positive cells was measured by flow cytometry (FACScan; Becton Dickinson). Acquired data were analyzed by the use of Cell Quest software [
23].
3.9. Detection of Bcl-2-Associated X Protein (Bax), B Cell Lymphoma 2 (Bcl2), and P53 Genes Expression by Reverse Transcriptase Real-Time–PCR
Control and treated cells were harvested, and total RNA was isolated using the RNeasy Mini Kit (Qiagen, Venlo, The Netherlands) following manufacturer’s protocol that encloses extra steps to remove genomic DNA. The quality and purity of isolated RNA were estimated spectrophotometrically at 260 nm, and the 260/280 nm ratios, respectively, using Nanodrop (Thermo Scientific, Waltham, MA, USA). 1 μg RNA from each sample was reverse-transcribed into cDNA using the Thermo Scientific cDNA Synthesis Kit (Promega, Leiden, The Netherlands).
RT-PCR primers for studied genes BAX, Bcl-2, and p53 were designed using Primer-BLAST software from the National Center for Biotechnology (Bethesda, MD, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a reference gene. CFX96 RT-PCR system (Bio-Rad, Hercules, CA, USA) was used to run RT-qPCR experiments using the following reaction mixture: 1 mM of forwarding/reverse primer (Eurogentec, Seraing, Belgium), 200 mM of dNTPs, and 0.75 U of Taq Polymerase (Roche). The thermal cycle was set using the following parameters: initial denaturation step at 94 °C for 5 min, 35 cycles of denaturation (60 s each, at 95 °C), annealing at 55 °C for 60 s, and an elongation step at 72 °C for 45 s, followed by final extra elongation step at 72 °C for 10 min [
24]. The sequences of studied primers and product size were depicted in
Table 7.
3.10. Statistical Methods
Data was analyzed using Statistical Package for Social Science software computer program version 26 (SPSS, Inc., Chicago, IL, USA). Quantitative parametric data were presented in mean and standard deviation while quantitative non-parametric data were presented in median (IQR). One-way analysis of variance (ANOVA) followed by post-hoc Tukey was used for comparing quantitative parametric data while Kruskal–Wallis followed by post-hoc Dunn’s was used for comparing quantitative non-parametric data. A log dose response curve was established to calculate IC50 for CIS and Schiff base complex in SCC and GMSCs groups by using graph pad prism version 8.0. p value less than 0.05 was considered statistically significant.
4. Discussion
Oral SCC is one of the most common cancers that remains noncurable with the existing therapies, and the survival of patients still has been a significant problem. Indeed, advanced oral SCC is chemotherapeutic and radiation-resistant by the current regimes [
25]. So, additional augmentations in the new chemotherapeutic agents for oral SCC might improve patient survival rates and outcomes. Indeed, the identification of novel therapeutic choices is an aim of interest.
The response rate of oral SCC to cytotoxic agents has not reached acceptable levels. Moreover, both the tumor cells’ resistance and dose-related toxicity stay two of the utmost significant problems in the chemotherapy of oral SCC [
26]. These drawbacks have pointed researchers to think about combination therapy, in which different chemotherapeutic agents are combined with other cytotoxic compounds that enable decreased chemoresistance and augmented anti-tumergenic effect. Among the available anticancer agents, cisplatin has been widely studied to be used in combination therapy. However, to design a well combinative chemotherapeutic regime, there must be a better focus on lower systemic toxicity along with enhanced cytotoxic effect.
Schiff bases have been discovered to show a wide variety of potential therapeutic purposes, including anticancer and antimicrobial effects. Schiff bases retain the high potential to inhibit carcinogenesis in various cancers and this reported effect can be enhanced with complexation. However, their antitumor activity mechanism is still in doubt [
27,
28,
29]. Meanwhile, new evolving data support the hopeful anticancer effect of Schiff base compounds against numerous types of cancer in vitro, like renal cell carcinoma [
12], prostate cancer [
30], hepatocellular carcinoma [
10], and colon cancer [
31]. On the other hand, its effect on oral SCC hasn’t been widely studied.
Therefore, the present study was designed to test the cytotoxic and chemosensitizing effects of heterodinuclear Cu(II)–Mn(II) Schiff base tetradentate complex on oral SCC to recognize its potential promising role in oral cancer therapy. We used heterodinuclear Cu(II)–Mn(II) Schiff base tetradentate complex alone and in combination with cisplatin against oral SCC cell line. We also used GMSCs as control.
MTT assay revealed the cytotoxic effect of the Schiff base and cisplatin on oral SCC. The IC50 value was calculated to be used for further experiments. Calculated IC50 values indicated less toxicity of Schiff base on both oral SCC and GMSCs. Moreover, Schiff base and cisplatin treatment led to the up-regulation of expression of p53 and Bax genes (pro-apoptotic), and down-regulation of Bcl2 (anti-apoptotic). These results explained data from flow cytometry, which indicated induction of apoptosis in treated cells compared to untreated controls.
Moreover, the combination of cisplatin with Schiff base demonstrated greater effects compared to sole treatment. Our data are in agreement with the data of Trávníček et al. [
32] who reported the cytotoxic effect of Schiff base ligand against ovarian carcinoma. Moreover, DNA cleavage activity and apoptosis of cancerous cells were previously reported as a mechanism Schiff base cytotoxic effect. Schiff base treatment alone and in combination with cisplatin compared to cisplatin alone caused more significant DNA damage and higher % of apoptotic cells arrested in the G0/G1 phase, as showed by flowcytometric analysis. These data are consistent with earlier studies recording that these complexes arrested the cell cycle in the G0/G1 phase and encouraged tumor cell apoptosis through a reactive oxygen species (ROS)-mediated mitochondrial pathway. Indeed, Schiff base cytotoxicity toward the HeLa cell lines was 1.9–3.5-fold more potent than cisplatin [
33].
It is usually accepted that initiation of apoptosis is the primary cytotoxic tool of most cancer chemotherapeutic agents, and deviations in the control of the apoptosis process can affect the sensitivity of malignant cells to multiple drugs. These chemotherapeutic drugs mediate their effect via modulating various cellular apoptotic proteins and regulators. Previously, various Schiff base metal complexes demonstrated cytotoxicity via triggering initiator and effector caspases. Both initiator and effector caspases play fundamental role in apoptotic process. Activated initiator caspases subsequently activate specific effector caspases leading to proteolytic degradation of intracellular proteins to mediate apoptosis [
27]. Moreover, p53 is a well-known regulator of apoptosis, and its variable expression has been involved as a reason for insensitivity or resistance of tumor cells [
34]. The p53 apoptotic pathway is frequently disturbed in most human cancer [
35].
P53-mediated apoptosis pathway comprises modifications of numerous executive components in apoptotic machines. One of them is Bax protein that functions as an apoptotic activator. The expression of the Bax gene is regulated by the tumor suppressor P53 but, this pro-apoptotic protein is inhibited by Bcl-2 that is localized to the outer membrane of mitochondria, as it plays a central role in supporting cellular survival and preventing the actions of pro-apoptotic proteins [
36]. Therefore, BAX/Bcl-1 balance is crucial for the process of cell death. Bulatov, 2018 et al. [
37] suggested that a Schiff base complex anti-proliferative effect can be both p53-dependent and independent. However, the presence of p53 exacerbates the cytotoxic effect of the complex. Herein, gene and protein expression analysis results showed that the used Schiff base complex could efficiently promote cell apoptosis via up-regulating the expression of P53 and Bax gene, caspase-3 and Bax proteins and down-regulating Bcl-2 gene and protein. These results are supported by a recent study by Zhang et al. [
38] who reported the cytotoxic effect of a Schiff base derivative on liver cancer cells via modulatory effect on apoptosis proteins.
Moreover, Song et al. [
39] studied the potential cytotoxic effects of three Schiff base complexes and reported specific cytotoxicity of these compounds to A549 cancer cells. The reported cytotoxic effect was mediated via the induction of DNA damage and inhibiting DNA synthesis. Similarly, our data demonstrated that Schiff base complex induced DNA damage in oral SCC, and this effect was enhanced on Schiff base complex cisplatin combination.