1. Introduction
Approximately 1.9 million cases of cancer are estimated to be diagnosed in 2023, with over 600,000 individuals expected to die from the disease. Brain and other nervous system tumors account for 3% of all cancer-related deaths in both sexes. Glioblastoma multiforme, the most common primary malignant brain tumor in adults, has an incidence rate of 3.26 per 100,000 populations. The term 'glioblastoma' is reserved for the IDH-wildtype diffuse gliomas grade 4, as defined by Louis et al. in 2021 [
1]. These tumors are characterized by rapid proliferation and poor differentiation. Genomic mutational studies, including those by The Cancer Genome Atlas (TCGA) Research Network in 2008, show that 80–90% of glioblastoma tumors harbor mutations affecting the PI3K, AKT, RAS, MAPK, p53, and/or RB signaling pathways. Glioblastomas are also highly angiogenic cancers with markedly increased VEGF expression, as noted by Reardon and Wen in 2015 [
2]. However, the therapeutic outcome following antiangiogenic therapy is highly variable in improving overall survival, according to studies by Chinot et al. in 2014 and Gilbert et al. in 2014 [
3,
4]. The current standard of care for glioblastoma patients, as outlined by Batash et al. in 2017 [
5] and the National Comprehensive Cancer Network (NCCN) in 2015, includes surgical resection, radiotherapy, and adjuvant chemotherapy with temozolomide. Despite recent advances in diagnosis and treatment, the overall survival for patients with glioblastomas is very poor, with a median overall survival of 13.5 months, as reported by Marenco-Hillembrand et al. in 2020 [
6] and Ostrom et al. in 2022 [
7]. Treatment outcomes in GBM patients are hindered by several factors, including high rates of
de novo and acquired resistance, incomplete surgical resection due to the tumor's highly infiltrative nature, low selective cytotoxicity, serious side effects, an immunosuppressive microenvironment, redundancy in aberrantly activated cell signaling pathways, and the challenge of effectively delivering therapeutics due to the blood–brain barrier. These challenges are highlighted in studies by Davis in 2016 [
8] and Reardon & Wen in 2015 [
2].
In recent years, there has been a surge of interest in exploring the therapeutic potential of medicinal plants for anticancer agents within the scientific community. Many conventional anticancer drugs are derived from bioactive constituents, including Vinca alkaloids (vinblastine and vincristine), cytotoxic podophyllotoxins, and Taxol [
9].
Nigella sativa Linn., commonly known as Blackseed or Black cumin, is an annual flowering plant native to South and Southwest Asia, and is prevalent in Northern Africa, the Middle East, and Southern Europe [
10]. It is one of the most promising and extensively studied medicinal plants for anticancer agents. Phytochemical studies have identified several bioactive compounds in
Nigella sativa, including sterols, saponins, phenolic compounds, alkaloids, novel lipid constituents, fatty acids, and volatile oils [
11]. The most pharmacologically active constituent of
Nigella Sativa's volatile oil is Thymoquinone (TQ) (2-methyl-5-isopropyl-1,4-benzoquinone) [
12,
13]. TQ is extracted from the essential oil of
Nigella Sativa seeds using supercritical fluid extraction [
14] and has been found to possess antimicrobial, antihistaminic, antidiabetic, anti-inflammatory, antioxidant, hypolipidemic, and anticancer properties [
15,
16,
17]. Several in vitro and in vivo studies have illustrated the cytotoxic effects of TQ on various cancer cell lines, including breast adenocarcinoma, leukemia, lung cancer, colorectal carcinoma, pancreatic cancer, osteosarcoma, prostate cancer, and glioblastoma [
17]. TQ affects multiple pathways and processes relevant to cancer, exhibiting anticancer activity through numerous mechanisms, specifically by showing selective antioxidant and oxidant activity, interfering with DNA structure, affecting carcinogenic signaling molecules/pathways, and immunomodulation [
18]. In vivo studies on mice have suggested that TQ can cross the blood-brain barrier due to its size and lipophilicity [
19]. Although some underlying mechanisms for the anticancer activities of TQ in glioblastoma cells have been identified, many potential pathways and targets remain to be fully elucidated. Research on the exact molecular pathways of its anticancer activities in glioblastoma cells is still in its early stages. Most research on TQ in glioblastoma cells has been conducted using the U-87 MG cell lines, with other glioma cell lines yet to be extensively studied. The aim of this study was to use RNA sequencing to examine changes in the expression of genes in A172 glioma cells in response to treatment with TQ at concentrations where significant cell death occurs. We then explored the pathways associated with those differentially expressed genes.
2. Materials and Methods
2.1. Cell Culture, Treatment and Cell Viability Assays
The glioblastoma cell line, A172, was purchased from the American Type Culture Collection (ATCC #CRL-1620) (Manassas, VA, USA). Cells were maintained in Dulbecco's modified Eagle medium (DMEM) (ATCC, Manassas, VA) containing 4,500 mg/L D-glucose, L-glutamine, and 110 mg\L sodium pyruvates in 75 or 175 cm2 cell culture flasks. The media was supplemented with 10% (v/v) Fetal bovine serum (FBS) (Atlas Biologicals, Fort Collins, CO). Additionally, an antibiotic-antimycotic mixture: of penicillin (10 000 U/mL), streptomycin (10mg/ mL), and amphotericin (25µg/mL) (Millipore Sigma Life Sciences, Burlington, MA) was also supplemented to the media. All cell lines were maintained in a humidified incubator at 37oC with 5% atmospheric CO2. For passaging the cells, trypsin-EDTA (ATCC, Manassas, VA) was used. 50mM stock solution of thymoquinone (Sigma Aldrich, MA) was prepared in filter sterilized DMSO (Millipore Sigma). Using the complete DMEM medium, suitable working concentrations were prepared from the stock solution. The final concentration of DMSO was 0.1% for all treatments. Cells were treated with different concentrations of thymoquinone ranging from 0 to 50 µM for 48 hours. After overnight attachment of cells, the media was removed and replaced with the corresponding volume of TQ-containing and untreated media. To test the effect of different concentrations of thymoquinone on cell viability, Toluidine blue staining Assay and Cell Counting Kit-8 was used.
2.2. Toluidine Blue Staining
A172 cells were seeded into 96 well plates and incubated overnight. 24 hours after incubation the media was removed, 100µl of thymoquinone-treated and untreated media was added to each well. Cells were treated with 0, 10, 25, and 50µM concentrations of TQ for 48 hours. Following the TQ treatments, the media was removed from each well. Cells were washed once with phosphate-buffered saline (PBS) (ATCC). 100µl of toluidine blue staining solution (1% toluidine blue (LabChem Inc, Zelienople, PA) and 1% borax (LabChem)) was added to each well and incubated for 20 minutes at room temperature. After 20-25 minutes, the staining solution was directly discarded to the sink. The remaining unbound solution was washed by immersing the plate in water. The plate was inverted and left to dry. The next day, the cells absorbance at 600nm was measured using SpectraMax iD3 Multi-Mode Microplate Reader (Molecular Devices, San Jose, CA).
2.3. Cell Counting Kit-8
Cell Counting Kit-8 (CCK-8) is a highly sensitive colorimetric assay widely used for the determination of a number of viable cells in cell viability and cytotoxicity assays. CCK-8 utilizes Dojindo's highly water-soluble tetrazolium salt, WST-8. The WST-8 assay was performed according to the manufacturer's protocol (Dojindo Laboratories, Gaithersburg, MD). A172 cells were seeded into 96 well plates at a density of not more than 5000 cells per well. Cells were incubated overnight for reattachment to the culture plate. The next day, the media was removed and replaced with fresh thymoquinone-treated and untreated media. Control, 10µM, 25µM, and 50µM of thymoquinone were used as treatment. After 48 hours of treatment, 10 µL of CCK-8 solution (Dojindo Laboratories) was added to each 96 wells and incubated for 3 hours at 37°C and 5% CO2. After incubation, the absorbance was measured at 450nm using SpectraMax iD3 Multi-Mode Microplate Reader (Molecular Devices).
2.4. RNA Extraction, Library Preparation, and RNA-Sequencing
A172 cells were seeded in 60mm culture dishes and incubated overnight. After 24 hours of incubation, the cells were treated with 25 and 50 µM concentrations of thymoquinone for 48 hours. Total RNA was extracted using Qiagen RNeasy Total RNA Isolation Kit (QIAGEN, Valencia, CA). The concentration and integrity of RNA samples were evaluated using a Nanodrop spectrophotometer (Thermo Fisher Scientific) and 0.7% % agarose gel electrophoresis. RNA was quantified using Qubit 3.0 (Thermo Fisher Scientific, USA). Then, the next-generation sequencing libraries were constructed as per the manufacturer's protocol ((NEBNext®Ultra™ RNA Library Prep Kit for Illumina®; Illumina, New England Biolabs Inc., Ipswich, MA, USA, Cat #E7770S/L, #E7775S/L). 1 µg of total RNA from each of the three biological replicates were used for the library preparation. The Poly(A) mRNA isolation was performed using NEBNext Poly(A) mRNA Magnetic Isolation Module (NEB #E7490). Similarly, NEBNext First-Strand Synthesis Reaction Buffer and NEBNext Random Primers mix were used for the fragmentation and priming of mRNA. First-strand cDNA was synthesized using NEBNext First Strand Synthesis Enzyme Mix, and the second-strand cDNA was synthesized using NEBNext Second Strand Synthesis Enzyme Mix. The double-stranded cDNA was purified using SPRIselect Beads. Thus, purified double-stranded DNA was treated with an End Prep Enzyme Mix to repair any broken or damaged ends and add a single A-base to the 3' end of the DNA. This was followed by the adapter ligation on both ends of cDNA. The size selection of the adapter-ligated DNA was performed using SPRIselect Beads to remove any DNA fragments that are too short or too long for efficient sequencing. PCR enrichment of size-selected DNA was performed using i5 and i7 primers. Purification of PCR products was performed using SPRIselect beads. The final library quality and insert size were determined using a bioanalyzer (Invitrogen, USA), and the library was quantified using a Qubit fluorometer (Invitrogen, USA). The library was diluted to 4nM concentration and sequenced using Illumina's NextSeq 500 platform with paired-end sequencing chemistry. The resulted image files in the BCL format were converted to FASTQ with 2x150 bp reads using the bcl2fastq tool (Illumina, USA).
2.5. Differential Gene Expression and Pathway Analysis
The raw data was obtained in FASTQ format. The quality of the raw data was assessed using the tool FastQC. For quality control, these raw reads were processed by Trimmomatic (v0.30) to remove low-quality bases (Quality Score <30) and those containing the adapter sequences (Bolger et al., 2014). Clean data obtained after quality trim was used for downstream analysis. The reference human genome sequences (GRCh38.p13) and the gene annotation files were then downloaded from gencode website (
https://www.gencodegenes.org/human/). STAR (version 2.7.3a) [
20] was used to index the reference human genome sequences in order to create the necessary data structures for efficient alignment. After creating the reference genome indexed file, the clean data were aligned to them via STAR aligner [
20]. In the next step, the aligned transcriptomic data in SAM format was converted into BAM format using SAMTools. The aligned BAM file was indexed using the SAMtools. The read count table was generated from the BAM alignment file and general feature format (GFF) of genome annotation using the HTSeq R package [
21]. The Differentially expressed genes (DEGs) among different experimental pair-wise combinations were identified using the DESeq2 [
22]. The DEGs were filtered based on the minimum Log2 Fold-Change (Log2FC) greater than 1 or less than -1 with a false discovery rate (FDR)⩽0.05. GO and Pathway Enrichment analyses were performed using ShinyGO [
23]. Similarly, heatmap was generated using statistical package pheatmap in RStudio, venn-diagram using Venny 2.1, and volcano plot was generated using online tool Galaxy Bioinformatics.
2.6. Real-time reverse transcription polymerase chain reaction (qRT-PCR)
Briefly, cells were treated with control, 10, 25, and 50µM concentrations of TQ for 48 hours. Total RNA was isolated and purified. Then, 500ng of total RNA was reverse transcribed into cDNA using random primers in a ImProm-II Reverse Transcription System (Promega, Madison, WI) following the manufacturer protocol. The RT reaction was carried out in a total volume of 20 µl. The quality and quantity of cDNA were assessed using a Nanodrop spectrophotometer (Thermo Fisher Scientific). cDNA from 27ng of RNA template and 100μM of each target genes primers was used. The mRNA expression level of the target gene was normalized to 18S rRNA mRNA expression. Fold change in the mRNA expression relative to the control was calculated using the RQ Study component of the ABI SDS v1.2.3 software and the comparative method based on 2-ΔΔCt values [
24].
2.7. Statistical Analysis
To test for significance, a one-way analysis of variance (ANOVA) was used. This test was used to determine whether there were statistically significant differences among the means of control and treatment groups. Data were presented as means + SEM. The confidence interval was 95%, and the P<0.05 was considered significant. For the multiple comparisons of the means, Tukey HSD post hoc test was conducted on R Studio.
4. Discussion
In our study investigating the impact of thymoquinone (TQ) on A172 glioma cell viability, we observed a dose-dependent inhibition of cell growth at higher concentrations, particularly after 48 hours of treatment. These results are consistent with existing literature, such as Ballout and Gali-Muhtasib, 2020 [
26], who reported similar time and concentration-dependent effects of TQ in neuroblastoma cells, and Racoma et al. (2013) [
27], who noted TQ's dose-dependent apoptotic induction in cancer cells. The dual role of TQ in modulating cell proliferation, as seen in our findings and echoed by Fatfat et al. (2021) [
28], highlights its complex biological activity. Our reliance on the WST-8 assay for its reliability, as also suggested by Eid et al. (2023) [
29], helped mitigate potential experimental errors common in assays like Toluidine Blue, as discussed by Adilovic et al. (2020) [
30]. Additionally, our observation of TQ's selective cytotoxicity aligns with Mokashi 2004 [
31], who emphasized its potential as a cancer therapeutic due to its ability to selectively target cancer cells.
In this study, TQ was found to inhibit the extracellular matrix-receptor (ECM-receptor) interaction pathway in A172 glioblastoma cells, affecting 8 differentially expressed genes. The ECM, which forms the cellular microenvironment and consists of glycoproteins, proteoglycans, and glycosaminoglycans [
32], is often overexpressed in glioma cells. Components such as hyaluronic acid, brevican, tenascin-C, fibronectin, thrombospondin, and specific integrins and receptors are known to promote cell adhesion and migration [
33]. These components, particularly HA and collagens, also act as barriers that limit drug diffusion and penetration into tumors [
34]. Additionally, collagen can inhibit T cell migration at tumor sites, impairing immune surveillance [
35]. TQ treatment's downregulation of several ECM components suggests potential therapeutic effects, such as decreased cell adhesion, impaired cancer cell migration, and hindered invasion through the basement membrane.
Further we noted, TQ inhibited the calcium signaling pathway in A172 glioblastoma cells, as evidenced by the downregulation of genes associated with this pathway following treatment with 25 and 50 µM TQ for 48 hours. Calcium signaling, crucial in controlling diverse cellular functions and linked with the progression of GBMs and other cancers [
36,
37], saw notable gene expression changes. CAMK2A, a key kinase in Ca2+-induced signaling and cell cycle progression, was significantly downregulated, aligning with findings by Colomer & Means (2007) [
38] and Takemoto-Kimura et al. (2017) [
39] on its role in proliferation. Studies by Wang et al. (2022) [
40] and Yu et al. (2021) [
41] further emphasize its impact on glioma cell functions. The TACR1 gene, encoding a receptor linked to various cancers [
42], also showed decreased expression, highlighting TQ's potential in cancer cell apoptosis. Moreover, the reduced expression of GRM1, involved in cell cycle arrest and apoptosis in glioma [
43,
44], and the downregulation of PLCG2, known for its overexpression in glioma and role in intracellular Ca2+ signaling [
45,
46], further underscore TQ's multifaceted impact on glioblastoma cell signaling and tumor progression.
In our study, KEGG pathway analysis indicated that higher concentrations of Thymoquinone (TQ) (50 µM) significantly downregulated genes in the PI3K-Akt signaling pathway in A172 glioblastoma cells, involving 18 differentially expressed genes like LPAR2, FGF10, COL9A3, PDGFB, IL2RB, IL7, PDGFRB, ITGB6, PPP2R2B, COL1A2, THBS3, PDGFD, GNG7, ERBB4, COL4A5, LAMA2, COL6A6, ITGA1. This pathway, crucial in processes like proliferation, differentiation, migration, metabolism, and survival, is often dysregulated in GBM due to RTK gene mutations and PI3K pathway activation. Genes like PDGFB, ERBB4, and IL-7, which play roles in activating PI3K-Akt signaling [
47,
48,
49,
50], were also downregulated. Interestingly, we observed the upregulation of RASD1, a gene that inhibits glioma cell migration and invasion by deactivating the AKT/mTOR signaling pathway [
51]. This might contribute to the observed inhibition of the AKT pathway. Additionally, PPP2R2B, often downregulated and methylated in GBM, was also downregulated in TQ-treated cells, correlating with shorter survival in GBM patients [
52]. These findings suggest TQ's potential as a therapeutic target in GBM by influencing key pathways and gene expressions.
In our study, we observed that 25 and 50 µM TQ treatments for 48 hours upregulated the P53 signaling pathway in A172 glioblastoma cells, without altering the level of p53 itself. This suggests TQ modulates the p53 pathway by influencing the expression of its downstream target genes. Specifically, TQ upregulated genes like PMAIP1 (NOXA), GADD45A, BBC3 (PUMA), and DUSP5. PMAIP1 and BBC3, transcription targets of p53 involved in DNA damage-induced apoptosis [
53,
54,
55,
56], are pro-apoptotic proteins of the Bcl-2 family, which, when activated, bind and inhibit anti-apoptotic proteins. The downregulation of the anti-apoptotic protein Bcl-XL was also noted. GADD45A, rapidly induced in response to DNA damage [
57,
58], and studies have shown its importance in apoptosis induction by anticancer agents [
59,
60,
61]. DUSP5, a dual-specificity phosphatase, deactivates protein kinases and its overexpression has been linked to suppressed growth in various cancer cells [
62]. The increase in proapoptotic proteins and decrease in anti-apoptotic proteins in TQ-treated cells suggest TQ may enhance mitochondrial membrane permeability, potentially activating the intrinsic pathway of apoptosis in glioblastoma cells.
In our study, TQ treatment of A172 glioblastoma cells with 25 and 50 µM concentrations for 48 hours significantly upregulated two candidate tumor suppressor genes: Sprouty RTJ Signaling Antagonist 4 (SPRY4) and Brain Expressed X-Linked 2 (BEX2). SPRY4, upregulated by 2.5-fold, is part of the Spry protein family, known to be repressed in GBM and often missing or deleted in gliomas [
63]. As a negative regulator of MAPK activation, its ectopic expression has been shown to inhibit proliferation and migration in GBM cells, suggesting its role as a tumor suppressor [
64]. BEX2, on the other hand, showed an upregulation by 3.2-fold in 50 µM TQ treatment and 2.5-fold in 25 µM treatment. Part of the BEX gene family, it was found to be silenced in U-87 and primary glioma cell lines, with its re-expression increasing sensitivity to chemotherapy-induced apoptosis and demonstrating tumor suppressor effects [
65]. However, some studies show contrasting results about BEX2, such as its high expression in glioma tissue and its role in inhibiting glioma cell migration and invasion by affecting β-catenin levels [
66]. These findings suggest that TQ may play a role in modulating tumor suppressor gene expression, contributing to its potential therapeutic effects in glioblastoma treatment.
In our study on A172 glioblastoma cells, TQ regulated the expression of key components and regulators of the Wnt signaling pathway. TQ increased the expression of Wnt Family Member 7B (Wnt7B) by 2.2-fold and decreased WNT6 expression by 1.6-fold. The Wnt pathway, known for its activation in glioblastoma, plays a crucial role in processes like cell proliferation, apoptosis, migration, and invasion. Studies have indicated varied expression levels of Wnt pathway components in gliomas, such as elevated WNT3A and 5A and decreased WNT7B [
67], and the oncogenic role of overexpressed WNT6 [
68]. In our study, Phophodiesterase 2A (PDE2A) was upregulated, a gene known to suppress Wnt/β-catenin signaling in glioma stem-like cells by modulating cAMP accumulation and GSK-3β phosphorylation [
69]. Additionally, TQ treatment led to downregulation of Sema3C, an overexpressed gene in most GBMs that activates the canonical Wnt pathway [
70]. Our findings suggest that TQ modulates Wnt signaling by altering the expression of its core components (WNT7B and WNT6) and regulators (PDE2A and Sema3C), thereby potentially downregulating this oncogenic pathway and contributing to its anticancer mechanism in vitro.
In our study on A172 glioblastoma cells, TQ treatment led to the upregulation of important genes like CHAC1 and DNER, known for their roles in GBM development. CHAC1, a cytosolic protein, typically downregulated in GBM cell lines, was identified as a key gene upregulated by TMZ treatment, enhancing glioma apoptotic death and inhibiting Notch3-mediated pathways [
71]. DNER, a noncanonical Notch ligand, was found to suppress glioma growth by inhibiting the oncogene TOR4A [
72] and hindered the growth and induced differentiation of GBM-derived neurospheres [
73]. Conversely, TQ treatment resulted in the downregulation of several genes overexpressed in GBM cells, including potential oncogenes like AEBP1, MIAT, GHR, LMO1, ELF3 [
74,
75,
76,
77,
78], and genes involved in tumor proliferation and migration such as EPHA4, COL3A1, PCDH10, ROBO1, ADAMTS5, PCDH18, ST8SIA1 [
79,
80,
81,
82,
83,
84]. Additionally, TQ significantly downregulated genes associated with poor GBM prognosis, including PCSK5, KCNC1, MXRA5, SEMA3C, MFAP2, MTERF2, KDM2B, FOXP2 [
85,
86,
87,
88,
89,
90,
91], indicating TQ's potential in targeting key molecular pathways in GBM.
Author Contributions
Conceptualization, G.R.H, M.S, W.D, U.K.R; methodology, R.P, G.R.H, P.N, C.S, U.K.R; software, R.P, P.N; validation, R.P, G.R.H; formal analysis, R.P, P.N, G.R.H; writing—original draft preparation, RP; writing—review and editing, P.N, U.K.R, G.R.H; supervision, G.R.H, P.N, U.K.R, W.D; project administration, G.R.H; funding acquisition, G.R.H, C. S, U. K. R. All authors have read and agreed to the published version of the manuscript.