Next Article in Journal
Glycan-Lectin Interactions as Novel Immunosuppression Drivers in Glioblastoma
Next Article in Special Issue
Asiatic Acid Attenuates Inflammation Induced by Salmonella via Upregulating LncRNA TVX1 in Microglia
Previous Article in Journal
Complex Elucidation of Cells-of-Origin in Pediatric Soft Tissue Sarcoma: From Concepts to Real Life, Hide-and-Seek through Epigenetic and Transcriptional Reprogramming
Previous Article in Special Issue
Berberine Suppresses Leukocyte Adherence by Downregulating CX3CL1 Expression and Shedding and ADAM10 in Lipopolysaccharide-Stimulated Vascular Endothelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone

1
Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering, Islamabad 54000, Pakistan
2
Department of Obstetrics and Gynecology, Yeditepe University, Istanbul 34280, Turkey
3
Food Science and Technology Program, BNU-HKBU United International College, Zhuhai 519087, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6311; https://doi.org/10.3390/ijms23116311
Submission received: 16 May 2022 / Revised: 29 May 2022 / Accepted: 30 May 2022 / Published: 5 June 2022

Abstract

:
Cancer is a life-threatening and multifaceted disease. Pioneering research works in the past three decades have mechanistically disentangled intertwined signaling networks which play contributory roles in carcinogenesis and metastasis. Phenomenal strides have been made in leveraging our scientific knowledge altogether to a new level of maturity. Rapidly accumulating wealth of information has underlined a myriad of transduction cascades which can be pharmaceutically exploited for cancer prevention/inhibition. Natural products serve as a treasure trove and compel interdisciplinary researchers to study the cancer chemopreventive roles of wide-ranging natural products in cell culture and preclinical studies. Experimental research related to thymoquinone has gradually gained momentum because of the extra-ordinary cancer chemopreventive multifunctionalities of thymoquinone. In this mini-review, we provide an overview of different cell signaling cascades reported to be regulated by thymoquinone for cancer chemoprevention. Essentially, thymoquinone efficacy has also been notably studied in animal models, which advocates for a rationale-based transition of thymoquinone from the pre-clinical pipeline to clinical trials.

1. Introduction

Deregulated cell signaling pathways play fundamental roles in re-shaping multiple facets of cancer, encompassing a wide range of functions that include the early initiation stage through to the metastatic stage, drug resistance, loss of apoptosis, and genetic/epigenetic inactivation [1,2,3,4]. Ground-breaking discoveries associated with the extra-ordinary pharmaceutical and medicinal values of natural products have catalyzed outstanding breakthroughs in drug development, consequently resulting in the rapid maturation of generalizable chemical platforms for the multi-targeting of previously undruggable oncogenic proteins [5,6,7,8,9].
Thymoquinone is isolated from Nigella sativa with a molecular weight of 164.2 g/mol. A series of cutting-edge research works have demonstrated the health-promoting and disease-ameliorating roles of thymoquinone [10,11,12,13,14]. Although different research teams have reviewed cancer chemopreventive role of thymoquinone [15,16,17,18,19], we summarize mechanistic insights gained through cell culture studies and xenografted mice. Herein, we provide a comprehensive overview of the fast-evolving field of pharmacological targeting of signaling pathways by thymoquinone in cancer and discuss the possibilities associated with targeting JAK/STAT, Wnt/β-catenin, PI3K/AKT/mTOR, NF-κB, and TRAIL-driven pathways for drug development in a wide range of oncology settings. We underline the knowledge gaps, identify key challenges, and make recommendations on how to rationally accelerate laboratory findings into clinically effective therapeutics.
For the framework of the mini-review, we extensively browsed PubMed using diverse keywords, particularly, “thymoquinone and cancer”, “thymoquinone and metastasis”, and “thymoquinone and signaling”.
In the upcoming sections, we will summarize thymoquinone-mediated regulation of cell signaling pathways in different cancers.

2. Regulation of JAK/STAT

A multimodule gene regulatory network mediates the complex process, involving signaling events and regulation of target genes, which modulates carcinogenesis and metastasis. The JAK/STAT pathway has been reported to be extensively involved in carcinogenesis and the spread of cancer cells to distant organs [20,21,22]. This section mainly deals with the thymoquinone-mediated inhibitory effects on the JAK/STAT pathway.
Thymoquinone inhibited the JAK2-mediated phosphorylation of STAT3 on the 727th serine residue in SK-MEL-28 cells. Importantly, levels of cyclin D1, D2, and D3 were reported to be reduced in STAT3-depleted SK-MEL-28 cells. Intraperitoneally administered thymoquinone caused tumor shrinkage in mice inoculated with SK-MEL-28 cells [23].
Gamma knife has been shown to be an effective treatment against brain metastasis from melanoma. Brain metastasis commonly occurs in patients suffering from metastatic malignant melanoma. Essentially, conversion of transient remissions to stable cures remains an overarching goal for clinical investigators of melanoma. Gamma knife and thymoquinone combinatorially enhanced the survival rates of C57BL/6J mice with intra-cerebral B16-F10 melanoma [24].
Furthermore, thymoquinone has also been reported to increase the survival rates in C57BL/6J mice with intracerebral B16-F10 melanoma cells. The JAK2/STAT3 pathway is inactivated by thymoquinone in B16-F10 melanoma cells. Collectively, these results clearly indicate that pharmacological targeting of melanoma brain metastasis by thymoquinone needs to be tested more comprehensively [25].
Renal cell carcinoma cells have high levels of anti-apoptotic proteins (Chae). It has also been noted that thymoquinone blocked the JAK2/STAT3-mediated upregulation of BCL-2, survivin, and cyclin D2 in Caki-1 cells (Figure 1). Intraperitoneal injections of thymoquinone hampered the growth of Caki-1 cell xenografts in nude mice [26].
Similarly, intraperitoneally administered thymoquinone impaired tumor growth rates in NSG mice xenotransplanted with epidermoid carcinoma A431 cells. Importantly, p53 is negatively regulated by MDM2. It was noted that thymoquinone increased p53 levels by simultaneous suppression in the level of MDM2 [27].
Thymoquinone inactivated the JAK2/STAT3 pathway in gastric HGC27 cancer cells. Thymoquinone also suppressed STAT3 target genes, such as survivin, VEGF, cyclin D, and BCL-2 (Figure 1). Intraperitoneally administered injections of thymoquinone induced tumor shrinkage in xenografted models [28].
Thymoquinone and cisplatin induced the regression of tumor xenografts. Importantly, p-STAT3 levels were noted to be profoundly reduced in the tumor tissues of thymoquinone and cisplatin-treated mice [29].
Evidence suggests that thymoquinone is efficient against acute myeloid leukemia. Thymoquinone chemically interacted with the active pockets of JAK2, STAT3, and STAT5 and inhibited their activities. Over the past two decades, following the discovery of the SOCS protein family, a wealth of knowledge has uncovered the functions and structures of SOCS proteins. SOCS proteins are negative-feedback regulators and a thymoquinone-mediated increase in SOCS proteins is necessary for inactivation of STAT proteins. Thymoquinone stimulated the expression of SHP-1, SOCS1, and SOCS3 in MV4–11 cells (Figure 1) [30].

3. Wnt/β-Catenin Signaling

With the advancements in sequencing technologies and detailed structural characterization of the cancer genome, it is apparent that WNT pathway mutations frequently occur in various cancers. β-Catenin is widely acclaimed as the principal transducer of canonical WNT signals to the nucleus. The orchestrated co-operation between cell surface mechanics and intracellular signaling has significant impacts on biological processes. In the absence of WNT ligands, β-catenin is phosphorylated by GSK3β, ubiquitylated by β-TrCP, and marked for degradation. The classical pathway is switched “on” upon binding of WNT ligands to frizzled receptors and LRP co-receptors, which results in stabilization and accumulation of β-catenin [31,32,33,34].
Thymoquinone dose-dependently inhibited nuclear accumulation of β-catenin [35]. Levels of β-catenin and Wnt/β-catenin target genes, such as c-Myc, matrix metalloproteinase-7, and Met, were found to be reduced in thymoquinone-treated bladder cancer cells. Importantly, β-catenin overexpression drastically abrogated the repressive effects of thymoquinone on the epithelial-to-mesenchymal transition by enhancing the levels of N-cadherin and vimentin. Intraperitoneally injected thymoquinone caused regression of tumor mass in xenografted mice. Moreover, pulmonary metastases models were generated by injections of T24-L-tagged luciferase for evaluation of the repressive effects of thymoquinone on distant metastasis. Moreover, bioluminescence imaging clearly indicated that pulmonary metastases were inhibited considerably by thymoquinone. Thymoquinone has been shown to significantly inhibit the foci of lung metastasis [35].
The MITF promoter present in close vicinity to the common downstream exon is known as the M promoter and expressed selectively in melanocytes [36]. Proteasomal degradation of β-catenin occurs through GSK3β-mediated phosphorylation at the serine-33, serine-37, serine-45, and threonine-41 residues of β-catenin. Studies have shown that phosphorylation at the Tyr-216 residue of GSK3β significantly enhances the enzymatic activity of GSK3β, whereas the phosphorylation at the 9th serine residue of GSK3β inactivates it. β-catenin has previously been reported to increase MITF expression. Therefore, strategic inactivation of β-catenin resulted in a decline in MITF levels. Thymoquinone dose-dependently reduced the expression of MITF and tyrosinase that was accompanied by decreased tyrosinase activity in B16F10 cells. Pre-treatment with LiCl resulted in an increase in the levels of p-GSK3β that led to the blockade of β-catenin degradation and increased the expression and activity of tyrosinase [36].
The generation of hybrids between conventional chemotherapeutic and bioactive natural products is an innovative approach to obtain effective anticancer compounds [37]. A hybrid of thymoquinone and 5-fluorouracil not only reduced β-catenin levels but also suppressed transcriptional activity of β-catenin in colorectal cancer cell lines. HCT116 control cells developed highly vascularized tumor masses, whereas hybrid-treated-tumor xenografts were significantly smaller in size. Interesting, there were signs of cellular proliferation in the xenografts combinatorically treated with thymoquinone and 5-fluorouracil, whereas hybrid-treated tumors did not show any sign of mitotic activity [37].
Intraperitoneally injected thymoquinone reduced the size and number of aberrant crypt foci and tumor multiplicities in a chemical-induced model of colorectal cancer [38]. Thymoquinone reduced polyp growth and selectively induced apoptosis. High doses of thymoquinone led to the translocation of β-catenin to the membrane and a reduction in the large polyps of APCMin mice [38].
It has been shown that the phosphorylation of GSK3β at serine-9 induced inactivation. Therefore, thymoquinone reduced p-GSK3β, β-catenin, and MMP2/MMP9 in Eca109 cells [39].

4. Regulation of UHRF1 by Thymoquinone

The ubiquitin-like protein containing PHD and RING fingers domains-1 (UHRF1) is a multi-domain-containing protein. UHRF1 maintains DNA methylation through the recruitment of DNA methyltransferase-1 to the replication forks in the S-phases of the cells.
Thymoquinone induced an auto-ubiquitination of UHRF1 through its RING domain [40]. The protein p73 is a functional and structural homologue of p53. P73 induced apoptosis in cancer cells. However, UHRF1 epigenetically inactivated p73 in Jurkat cells. However, thymoquinone-mediated auto-ubiquitination of UHRF1 caused a sharp increase in the expression of p73 in Jurkat cells [40].
UHRF1 binds to the inverted CCAAT domain in the promoter region of TXNIP and inhibits its expression via CpG methylation [41]. UHRF1 knockdown inhibited UHRF1 binding to the promoter of TXNIP and enhanced TXNIP expression through promoter demethylation in HeLa cells. Levels of ubiquitin-specific protease-7 (USP7) were noted to be enhanced in HPV16 E6/E7-overexpressing cells. USP7 efficiently stabilized UHRF1 and epigenetically inactivated TXNIP [41].
Likewise, HPV E6/E7 caused a marked increase in the levels of USP7 and UHRF1. UHRF1 effectively inactivated gelsolin in HeLa cells, whereas thymoquinone induced the expression of gelsolin and promoted apoptotic death in cancer cells [42].

5. PI3K/AKT/mTOR

The PI3K/AKT/mTOR pathway has long been an attractive target in molecular oncology. Research teams have focused on the pharmacological targeting of key components of this signaling network [43,44,45,46,47].
Thymoquinone dose-dependently reduced the levels of p-AKT (threonine-308), p-AKT (serine-473), p-mTOR1, and p-mTOR2 in gastric cancer cells. Thymoquinone inhibited the colony formation and invasive capacities of gastric cancer cells [48].
Thymoquinone alone and with pre-treatment significantly reduced p-AKT in BxPC-3, AsPC-1, and PANC-1 cells [49]. Thymoquinone pre-treatment markedly impaired gemcitabine-mediated increases in the phosphorylation of mTOR and S6 in pancreatic cancer cells. More importantly, thymoquinone and gemcitabine induced shrinkage of the primary tumors in the orthotopic cancer models of PANC-1 cells. Pre-treatment with thymoquinone led to marked suppression in p-AKT, p-mTOR, and p-S6 in tumor tissues of xenografted mice [49].
AMPK activates autophagy by inhibition of mTOR [50]. The inhibition of mTORC1 increased autophagy, whereas mTORC1 activation caused deactivation of autophagy. Thymoquinone caused an increase in the level of p-AMPK in 786-O and ACHN cells, while levels of p-mTOR and p-S6K were reduced. There was a significant decline in the number of metastatic nodules in the lungs of thymoquinone-treated mice [50].
Thymoquinone effectively inhibited the PI3K/AKT/mTOR pathway independently and in combination with 5-fluorouracil and active vitamin D3 in colorectal cancer cells [51].

6. NF-κb Activity in Carcinogenesis: Paradoxical Roles of Thymoquinone

Learning more about the complicated mechanisms of NF-κB regulation can be advantageous in the design and development of better therapeutic approaches to target versatile transcriptional factor in different types of cancers. NF-κB functions are controlled tightly by several regulatory proteins, and a disruption of this process has been associated with carcinogenesis and metastasis [52,53,54,55]. NF-κB provides a mechanistic linkage between cancer and inflammation and is a main regulator controlling the capability of malignant cells to trigger pro-survival signaling and resist apoptotic cell death.
Thymoquinone stimulated the expression levels of miR-603 in MDA-MB-436 and MDA-MB-231 cancer cells. NF-κB transcriptionally repressed miR-603. Importantly, miR-603 directly targeted eEF-2K and inhibited the proliferation and invasive capacities of breast cancer cells. Intravenously administered thymoquinone-loaded liposomal nanoparticles impaired tumor growth in mice orthotopically implanted with MDA-MB-231 and MDA-MB-436 cancer cells [56].
Thymoquinone and bortezomib combinatorially induced tumor shrinkage in mice subcutaneously implanted with U266 cells. NF-κB activity was significantly reduced in the tumor tissues of mice xenografted with multiple myeloma cells [57].
There was a significant decrease in the ratio of p-NF-κB/NF-κB in tumor xenografts from mice combinatorially treated with thymoquinone and cisplatin [58]
Thymoquinone inhibited NF-κB-mediated activity and prevented cancer progression [59,60].
Cancer-promoting role of Thymoquinone: Prolonged thymoquinone treatment results in an increase in NF-κB reporter activities and induced a two-fold rise in volume of the ascites [61].
ID8-NGL mouse ovarian cancer cells stably expressing NF-κB have been investigated to analyze the cancer chemopreventive effects of thymoquinone. ID8-NGL cells were intraperitoneally injected into a C57BL/6 rodent model. Prolonged treatment of thymoquinone (30 days) resulted in an increase in the activity of NF-κB in tumors. Moreover, prolonged treatment of thymoquinone induced pro-tumor M2-like macrophages within the tumor microenvironments. Thymoquinone triggered an increase in the infiltration of macrophages. Thymoquinone caused an overall increase in the expression of IL-1β and TNFα in macrophages and VEGF in ascites fluid. M2-like macrophages produced high concentrations of signaling molecules such as TNFα, which increased the activity of NF-κB in the tumor tissues, consequently resulting in drug resistance in a sub-population of cancer cells [62].

7. Chemokine Ligand-Driven Signaling

Thymoquinone effectively inhibited NF-κB-mediated transcriptional upregulation of CXCR4 [63]. Importantly, thymoquinone caused a marked reduction in the number of colonies metastasized to secondary sites. Moreover, CXCR4-expressing MDA-MB-231 cancer cells showed a tendency to migrate towards a CXCL12-expressing microenvironment in mice intracardially implanted with MDA-MB-231 cells. Thymoquinone treatment led to significant suppression of the metastatic colonization of breast cancer cells into the bones (Figure 2). Moreover, there was an evident reduction in metastatic sites, such as the bone marrow of the tibiae and femora, as well as the mandibles. Similarly, thymoquinone-mediated anti-metastatic effects were also observed in the lungs and brain tissues [63].
CXCL12 induced a physical association between CXCR4 and CD45 in multiple myeloma cells [64]. Multiple CD45-silenced myeloma cells completely lost their migratory potential in response to CXCL12. Thymoquinone decreased the surface expression of CXCR4 on multiple myeloma cells and CXCL12-mediated CXCR4-CD45 interactions [64].

8. Regulation of TRAIL-Mediated Signaling by Thymoquinone

The tumor necrosis factor-related apoptosis ligand (TRAIL) triggered apoptotic death through engagement of the death receptors DR4 and DR5 [65,66,67,68,69,70].
Thymoquinone effectively enhanced TRAIL-mediated DNA damage in HepG2 cells. Thymoquinone and TRAIL synergistically reduced the levels of XIAP (X-linked inhibitor of apoptosis), c-FLIP, BCL-2, cIAP1, and cIAP2 in HepG2 cancer cells [71]. Different studies have shown that Thymoquinone stimulates the expression of death receptors for activation of intracellular apoptotic signaling in HepG2 cells. Moreover, there is sufficient evidence for TRAIL-mediated activation of NF-κB in different cancers. However, thymoquinone efficiently reduced the levels of NF-κB [72,73].
Activated B-cell lymphoma (ABC), a subtype of diffuse large B-cell lymphoma (DLBCL), has a poor survival rate. Thymoquinone markedly reduced serine-32-phosphorylated IκBα. The phosphorylation of IκBα led to the release of NF-κB and the consequent activation of NF-κB-target genes [74].
Thymoquinone time-dependently decreased BCL-2 levels and simultaneously enhanced BAX levels [75]. Thymoquinone promoted the release of mitochondrially located cytochrome c to the cytosol in primary effusion lymphoma (PEL) cell lines (BC1 and BC3). N-acetyl cysteine caused a blockade of conformational changes in BAX protein. Essentially, thymoquinone induced structural re-orientation necessary for the activation of BAX. Thymoquinone-mediated ROS accumulation triggered conformational changes in BAX that sequentially resulted in the activation of the mitochondrial apoptotic pathway. Thymoquinone effectively increased the release of cytochrome c into the cytosol [75].

9. Regulation of EMT by Thymoquinone

The epithelial-to-mesenchymal transition (EMT)-associated reprogramming of cells not only showcases fundamental changes in different regulatory networks but also informs us about an intricate interplay that exists between them. Deregulation of a controlled epithelial balance is triggered by alterations in several regulatory layers. Our rapidly evolving conceptualization of the genetic evolution of metastatic diseases has enabled us to decode variability in the therapeutic vulnerabilities of primary and secondary/metastatic tumors [76,77,78]. Excitingly, cutting-edge and seminal researches have shed light on the routes to and temporal patterns of metastatic colonization and produced mechanistic insights into the complex barcodes which underlie metastasis [79,80].
Myrtucommulone-A and thymoquinone notably reduced the levels of vimentin, Slug, and N-cadherin, whereas levels of E-cadherin were found to be enhanced [81]. Thymoquinone downregulated TWIST1 and ZEB1 and simultaneously upregulated E-cadherin in SiHa and CaSki cell lines [82].

10. Regulation of the MAPK Pathway by Thymoquinone:

The MAPK transduction pathway encompasses a cascade of phosphorylation events involving three functionally active key kinases, namely RAF, MEK, and ERK [83,84,85,86,87,88,89,90]. Importantly, this exciting kinase cascade presents unique prospects for the design and development of novel and effective cancer therapeutics.
Thymoquinone induced an increase in the phosphorylated levels of JNK, p38-MAPK, and ERK. Thymoquinone-induced ROS enhanced the phosphorylation of p38-MAPK in MCF-7 cells. Thymoquinone-mediated apoptosis was impaired in p38-MAPK silenced cancer cells [91].
It had previously been shown that the transfection of cells with the non-phosphorylatable mutants T212A caused an increase in p-PAK(threonine-423) and enhanced apoptotic death [92]. Similarly, an increase in apoptosis was noted in cells transfected with kinase-dead K299R mutants and PAK1 siRNAs. The thymoquinone-induced activation of ERK1/2 is inhibited when PAK (threonine-423) is phosphorylated maximally. Secondly, the combination of IPA-3 (PAK inhibitor) with thymoquinone led to a decrease in the phosphorylation of threonine-423, accompanied by a significant rise in p-ERK1/2 levels that highlighted an early activation of the MEK-ERK cascade. Moreover, the lack of inhibition of p-PAK1 (threonine-423) is associated closely with a decrease in pro-survival ERK1/2 activation and a considerable increase in apoptotic cell death [92].
FR180204 (ERK inhibitor) significantly reduced the viability of thymoquinone and docetaxel-treated cancer cells [93]. Collectively, these findings indicate that ERK inhibition/inactivation reduced the viability of cancer cells.
Thymoquinone inhibited the proliferation, migration, and invasion of A549 cells by inactivating the ERK1/2 signaling cascade [94]. ERK and JNK activation protected DLD-1 cells from thymoquinone-induced oxidative stress and apoptotic cell death [95]. Therefore, a combination of thymoquinone with ERK and JNK inhibitors can efficiently enhance the cancer-inhibitory effects of thymoquinone.

11. Regulation of microRNAs by Thymoquinone

The discovery of microRNAs has altered our perception of non-coding RNAs from ‘junk’ transcriptional products to functional regulatory molecules [96,97,98].
PD-L1 is directly targeted by hsa-miR-877-5p. Thymoquinone promoted hsa-miR-877-5p-driven targeting of PD-L1 in T24 and 5637 bladder cancer cells. miR-877-5p inhibitors enhanced the migratory capacity of bladder cancer cells and weakened thymoquinone-induced inhibitory effects on the invasive potential of cancer cells. Thymoquinone and cisplatin induced tumor retrogression in mice inoculated with T24 cancer cells. hsa-miR-877-5p levels were found to be enhanced in tumor xenografts in mice inoculated with T24 cells. Moreover, thymoquinone profoundly suppressed the number of pulmonary metastatic nodules in mice injected with bladder cancer cells [99].
Src family kinases are nonreceptor tyrosine kinases involved in the modulation of many signaling cascades. Thymoquinone markedly reduced the levels of eEF-2K and suppressed the phosphorylated levels of Src and FAK [56]. Knockdown of eEF-2K caused significant inhibition of the migratory and invasive potential of MDA-MB-436 and MDA-MB-231 cancer cells. Importantly, there was a significant reduction in the levels of p-Src, p-AKT, p-FAK, and p-EF2 in MDA-MB-436 and MDA-MB-231 cancer cells. Thymoquinone significantly upregulated the expression of miR-603 in breast cancer cells. Studies have shown that miR-603 directly targets eEF-2K in cancer cells. The inhibition of NF-κB significantly enhanced miR-603 expression. Systemically administered thymoquinone effectively inhibited tumor growth in experimental mice implanted orthotopically with MDA-MB-231 and MDA-MB-436 cancer cells. In particular, levels of eEF-2K were found to be reduced in the tumor tissues of xenografted mice [56].
Thymoquinone and doxorubicin worked effectively and stimulated the levels of miR-375 and miR-16 in HepG2 and Huh7 cancer cells. Both miR-375 and miR-16 induced an increase in caspase-3 and a simultaneous reduction in the levels of BCL-2 [100].
Thymoquinone-loaded, hyaluronic-acid-conjugated Pluronic® P123- and F127-co-polymer nanoparticles have been shown to be efficient against triple-negative breast cancer cells [101]. Thymoquinone nanoparticles induced disruption of the stress fibers by downregulating Rho and Rac1 in MDA-MB-231 cancer cells. These nanoparticles stimulated the expression of the tumor suppressor miR-361 in MDA-MB-231 cancer cells. Importantly, miR-361 directly targeted Rac1 and Rho. Thymoquinone-loaded nanoparticles markedly suppressed the number of pulmonary metastatic nodules in mice orthotopically injected with 4T1 cancer cells [101].
Transferrin-decorated thymoquinone-loaded PEG-PLGA nanoparticles triggered the upregulation of miRNA-16 and miRNA-34a via p53 in H1299 cancer cells [82]. Thymoquinone-loaded nanoparticles reduced the migration of p53 (wild-type) expressing lung cancer cells. These nanoparticles induced shrinkage of the tumor mass in mice injected with A549 cancer cells [102].
Likewise, PEGylated thymoquinone nanoparticles have also been demonstrated to stimulate the expression of miR-34a through p53 in cancer cells [103]. miR-34a directly targeted Rac1, followed by actin depolymerization. Thereafter, the depolymerization of actin further disrupted the actin cytoskeleton, which significantly reduced filopodia and lamellipodia formation on cell surfaces, thus retarding the migration of cancer cells. PEGylated thymoquinone nanoparticles significantly suppressed tumor weight and tumor volume in tumor-bearing mice. These nanoparticles efficiently enhanced the levels of catalase and superoxide dismutase (SOD) in tumor-bearing mice. PEGylated thymoquinone nanoparticles prevented oxidative stress, which further ameliorated the overall systemic toxicities [103].

12. Metastasis Inhibitory Role of Thymoquinone: Animal Model Studies

TQFL12, a new derivative, is more effective compared to thymoquinone. TQFL12 caused shrinkage of tumor xenografts in mice orthotopically implanted with 4T1 cancer cells [104].
Tristetraprolin (TTP) is a well-known AU-rich element-binding protein. TTP has been demonstrated to exert tumor-suppressive effects [105]. MUC4 was destabilized by direct binding of tristetraprolin to ARE in 3’UTR of MUC4 mRNA. Thymoquinone treatment reduced colony formation. However, the colony-forming ability of TTP-silenced cancer cells was found to be considerably enhanced. Thymoquinone treatment or TTP overexpression significantly reduced cancer metastasis and combinatorial treatment synergistically reduced pulmonary metastatic nodules [105].
Indirubin-3-monoxime and thymoquinone severely hampered tumor growth in subcutaneous A549 xenograft models. Levels of p-AKT and p-mTOR were reduced in the tumor tissues. Both individual and combinatorial treatments caused an increase in caspase-3 and p53 levels in the tumor tissues [106].
Thymoquinone suppressed VEGF-mediated activation of ERK. However, thymoquinone did not inactivate VEGFR2. Thymoquinone abolished tumor growth in mice subcutaneously transplanted with PC3 cancer cells. Thymoquinone significantly inhibited tumor angiogenesis as it effectively reduced the number of blood vessels in the tumor mass [107].
The constitutive activation of NLRP3 results in auto-inflammation characterized by sustained systemic and local inflammation mediated by interleukin-1β [108]. Glyburide is an inhibitor of NLRP3 inflammasomes. Glyburide blocks NLRP3 activation and secretion of IL-18 and IL-1β. Thymoquinone significantly decreased the expression of NLRP3 inflammasomes in B16F10 and A375 melanoma cells. Inflammasomes elicited the proteolytic maturation and secretion of IL-1β and IL-18 through caspase-1. Thymoquinone decreased the proteolytic cleavage of pro-caspase-1 in B16F10 and A375 melanoma cells. LPS/ATP are NLRP3 inflammasome activators. LPS/ATP triggered the activation of NLRP3 inflammasomes, including caspase-1 activities, which enhanced the secretion of IL-18 and IL-1β, consequently resulting in an enhanced migratory ability of melanoma cells. There was a notable reduction in macroscopic metastatic nodules on the surface of the lungs in C57BL/6 mice injected with B16F10 melanoma cells [108].

13. Concluding Remarks

Increasingly, it is being acclaimed that the strategy of conducting both forward translation (the implementation of scientific discoveries into clinical practice) as well as reverse translation (the process of elucidation of the underlying mechanisms of clinical observations) significantly reinforces the capability of interdisciplinary researchers to design and develop highly efficient anticancer treatment strategies. Recent advancements in genomic technologies and rapidly evolving large gene expression datasets have enabled researchers to sharply resolve the gene signatures that characterize malignant phenotypes. On the translational forefronts, pioneering studies over the past two decades have spurred the acclamation of thymoquinone as an effective cancer chemopreventive agent. Certain hints have emerged which indicate thymoquinone-mediated inactivation of the SHH/GLI and TGFβ/SMAD pathways [109,110]. Furthermore, thymoquinone worked effectively in combination with vitamin D3 and 5-fluorouracil and increased the expression of TGFβ and SMAD4 in chemical-induced cancer models [111,112]. Additionally, it will be exciting to explore the regulation of long non-coding RNAs and circular RNAs by thymoquinone. These aspects will add and enrich various aspects of molecular oncology. However, the prolonged administration of thymoquinone has an oncogenic role in mice xenografted with ovarian cancer cells. Therefore, these aspects have to be kept in consideration, mainly in the context of ovarian cancer. Collectively, the thymoquinone-mediated regulation of oncogenic pathways is encouraging and advocates its pharmacological significance. Thymoquinone has been unraveled to a greater extent and future studies must converge on rationally designed clinical trials for a better analysis of thymoquinone in cancer chemoprevention.

Author Contributions

Conceptualization, A.A.F. and B.X.; methodology, A.A.F.; software, R.A.; resources, A.A.F.; writing—original draft preparation, A.A.F. and R.A.; writing—review and editing, B.X.; funding acquisition, B.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by BNU-HKBU United International College (grant number: R202107), and the APC was funded by BNU-HKBU United International College.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Puisieux, A.; Brabletz, T.; Caramel, J. Oncogenic roles of EMT-inducing transcription factors. Nat. Cell Biol. 2014, 16, 488–494. [Google Scholar] [CrossRef] [PubMed]
  3. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84. [Google Scholar] [CrossRef] [PubMed]
  4. Valastyan, S.; Weinberg, R.A. Tumor metastasis: Molecular insights and evolving paradigms. Cell 2011, 147, 275–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hamann, L.G. Synthetic strategy: Natural products on demand. Nat. Chem. 2014, 6, 460–461. [Google Scholar] [CrossRef]
  6. Mann, J. Natural products in cancer chemotherapy: Past, present and future. Nat. Rev. Cancer 2002, 2, 143–148. [Google Scholar] [CrossRef]
  7. Hann, M.M.; Keserü, G.M. Finding the sweet spot: The role of nature and nurture in medicinal chemistry. Nat. Rev. Drug Discov. 2012, 11, 355–365. [Google Scholar] [CrossRef]
  8. Keserü, G.M.; Makara, G.M. The influence of lead discovery strategies on the properties of drug candidates. Nat. Rev. Drug Discov. 2009, 8, 203–212. [Google Scholar] [CrossRef]
  9. Lombardino, J.G.; Lowe, J.A., III. The role of the medicinal chemist in drug discovery—Then and now. Nat. Rev. Drug Discov. 2004, 3, 853–862. [Google Scholar] [CrossRef]
  10. Harphoush, S.; Wu, G.; Qiuli, G.; Zaitoun, M.; Ghanem, M.; Shi, Y.; Le, G. Thymoquinone ameliorates obesity-induced metabolic dysfunction, improves reproductive efficiency exhibiting a dose-organ relationship. Syst. Biol. Reprod. Med. 2019, 65, 367–382. [Google Scholar] [CrossRef]
  11. Karandrea, S.; Yin, H.; Liang, X.; Slitt, A.L.; Heart, E.A. Thymoquinone ameliorates diabetic phenotype in diet-induced obesity mice via activation of SIRT-1-dependent pathways. PLoS ONE 2017, 12, e0185374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Badr, G.; Mahmoud, M.H.; Farhat, K.; Waly, H.; Al-Abdin, O.Z.; Rabah, D.M. Maternal supplementation of diabetic mice with thymoquinone protects their offspring from abnormal obesity and diabetes by modulating their lipid profile and free radical production and restoring lymphocyte proliferation via PI3K/AKT signaling. Lipids Health Dis. 2013, 12, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Alshahrani, S.; Anwer, T.; Alam, M.F.; Ahmed, R.A.; Khan, G.; Sivakumar, S.M.; Shoaib, A.; Alam, P.; Azam, F. Effect of thymoquinone on high fat diet and STZ-induced experimental type 2 diabetes: A mechanistic insight by in vivo and in silico studies. J. Food Biochem. 2021, e13807. [Google Scholar] [CrossRef] [PubMed]
  14. El-Shemi, A.G.; Kensara, O.A.; Alsaegh, A.; Mukhtar, M.H. Pharmacotherapy with thymoquinone improved pancreatic β-cell integrity and functional activity, enhanced islets revascularization, and alleviated metabolic and hepato-renal disturbances in streptozotocin-induced diabetes in rats. Pharmacology 2018, 101, 9–21. [Google Scholar] [CrossRef] [PubMed]
  15. Khan, M.A.; Younus, H. Thymoquinone shows the diverse therapeutic actions by modulating multiple cell signaling pathways: Single drug for multiple targets. Curr. Pharm. Biotechnol. 2018, 19, 934–945. [Google Scholar] [CrossRef] [PubMed]
  16. Majdalawieh, A.F.; Fayyad, M.W.; Nasrallah, G.K. Anti-cancer properties and mechanisms of action of thymoquinone, the major active ingredient of Nigella sativa. Crit. Rev. Food Sci. Nutr. 2017, 57, 3911–3928. [Google Scholar] [CrossRef]
  17. Schneider-Stock, R.; Fakhoury, I.H.; Zaki, A.M.; El-Baba, C.O.; Gali-Muhtasib, H.U. Thymoquinone: Fifty years of success in the battle against cancer models. Drug Discov. Today 2014, 19, 18–30. [Google Scholar] [CrossRef]
  18. Banerjee, S.; Padhye, S.; Azmi, A.; Wang, Z.; Philip, P.A.; Kucuk, O.; Sarkar, F.H.; Mohammad, R.M. Review on molecular and therapeutic potential of thymoquinone in cancer. Nutr. Cancer 2010, 62, 938–946. [Google Scholar] [CrossRef]
  19. Gali-Muhtasib, H.; Roessner, A.; Schneider-Stock, R. Thymoquinone: A promising anti-cancer drug from natural sources. Int. J. Biochem. Cell Biol. 2006, 38, 1249–1253. [Google Scholar] [CrossRef]
  20. Thomas, S.J.; Snowden, J.A.; Zeidler, M.P.; Danson, S.J. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br. J. Cancer 2015, 113, 365–371. [Google Scholar] [CrossRef] [Green Version]
  21. Recio, C.; Guerra, B.; Guerra-Rodríguez, M.; Aranda-Tavío, H.; Martín-Rodríguez, P.; de Mirecki-Garrido, M.; Brito-Casillas, Y.; García-Castellano, J.M.; Estévez-Braun, A.; Fernández-Pérez, L. Signal transducer and activator of transcription (STAT)-5: An opportunity for drug development in oncohematology. Oncogene 2019, 38, 4657–4668. [Google Scholar] [CrossRef] [PubMed]
  22. Miklossy, G.; Hilliard, T.S.; Turkson, J. Therapeutic modulators of STAT signalling for human diseases. Nat. Rev. Drug Discov. 2013, 12, 611–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Raut, P.K.; Lee, H.S.; Joo, S.H.; Chun, K.S. Thymoquinone induces oxidative stress-mediated apoptosis through downregulation of Jak2/STAT3 signaling pathway in human melanoma cells. Food Chem. Toxicol. 2021, 157, 112604. [Google Scholar] [CrossRef] [PubMed]
  24. Hatiboglu, M.A.; Kocyigit, A.; Guler, E.M.; Akdur, K.; Khan, I.; Nalli, A.; Karatas, E.; Tuzgen, S. Thymoquinone enhances the effect of gamma knife in B16-F10 melanoma through inhibition of phosphorylated STAT3. World Neurosurg. 2019, 128, e570–e581. [Google Scholar] [CrossRef] [PubMed]
  25. Hatiboglu, M.A.; Kocyigit, A.; Guler, E.M.; Akdur, K.; Nalli, A.; Karatas, E.; Tuzgen, S. Thymoquinone induces apoptosis in B16-F10 melanoma cell through inhibition of p-STAT3 and inhibits tumor growth in a murine intracerebral melanoma model. World Neurosurg. 2018, 114, e182–e190. [Google Scholar] [CrossRef] [PubMed]
  26. Chae, I.G.; Song, N.Y.; Kim, D.H.; Lee, M.Y.; Park, J.M.; Chun, K.S. Thymoquinone induces apoptosis of human renal carcinoma Caki-1 cells by inhibiting JAK2/STAT3 through pro-oxidant effect. Food Chem. Toxicol. 2020, 139, 111253. [Google Scholar] [CrossRef] [PubMed]
  27. Park, J.E.; Kim, D.H.; Ha, E.; Choi, S.M.; Choi, J.S.; Chun, K.S.; Joo, S.H. Thymoquinone induces apoptosis of human epidermoid carcinoma A431 cells through ROS-mediated suppression of STAT3. Chem. Biol. Interact. 2019, 312, 108799. [Google Scholar] [CrossRef]
  28. Zhu, W.Q.; Wang, J.; Guo, X.F.; Liu, Z.; Dong, W.G. Thymoquinone inhibits proliferation in gastric cancer via the STAT3 pathway in vivo and in vitro. World J. Gastroenterol. 2016, 22, 4149–4159. [Google Scholar] [CrossRef]
  29. Hu, X.; Ma, J.; Vikash, V.; Li, J.; Wu, D.; Liu, Y.; Zhang, J.; Dong, W. Thymoquinone augments cisplatin-induced apoptosis on esophageal carcinoma through mitigating the activation of JAK2/STAT3 pathway. Dig. Dis. Sci. 2018, 63, 126–134. [Google Scholar] [CrossRef]
  30. Al-Rawashde, F.A.; Johan, M.F.; Taib, W.R.W.; Ismail, I.; Johari, S.A.T.T.; Almajali, B.; Al-Wajeeh, A.S.; Nazari Vishkaei, M.; Al-Jamal, H.A.N. Thymoquinone inhibits growth of acute myeloid leukemia cells through reversal SHP-1 and SOCS-3 hypermethylation: In vitro and in silico evaluation. Pharmaceuticals 2021, 14, 1287. [Google Scholar] [CrossRef]
  31. Korinek, V.; Barker, N.; Morin, P.J.; van Wichen, D.; de Weger, R.; Kinzler, K.W.; Vogelstein, B.; Clevers, H. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC−/− colon carcinoma. Science 1997, 275, 1784–1787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Morin, P.J.; Sparks, A.B.; Korinek, V.; Barker, N.; Clevers, H.; Vogelstein, B.; Kinzler, K.W. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 1997, 275, 1787–1790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Suzuki, H.; Watkins, D.N.; Jair, K.W.; Schuebel, K.E.; Markowitz, S.D.; Chen, W.D.; Pretlow, T.P.; Yang, B.; Akiyama, Y.; Van Engeland, M.; et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat. Genet. 2004, 36, 417–422. [Google Scholar] [CrossRef] [PubMed]
  34. Behrens, J.; Jerchow, B.A.; Würtele, M.; Grimm, J.; Asbrand, C.; Wirtz, R.; Kühl, M.; Wedlich, D.; Birchmeier, W. Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science 1998, 280, 596–599. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, M.; Du, H.; Wang, L.; Yue, Y.; Zhang, P.; Huang, Z.; Lv, W.; Ma, J.; Shao, Q.; Ma, M.; et al. Thymoquinone suppresses invasion and metastasis in bladder cancer cells by reversing EMT through the Wnt/β-catenin signaling pathway. Chem. Biol. Interact. 2020, 320, 109022. [Google Scholar] [CrossRef] [PubMed]
  36. Jeong, H.; Yu, S.M.; Kim, S.J. Inhibitory effects on melanogenesis by thymoquinone are mediated through the β-catenin pathway in B16F10 mouse melanoma cells. Int. J. Oncol. 2020, 56, 379–389. [Google Scholar] [CrossRef]
  37. Ndreshkjana, B.; Çapci, A.; Klein, V.; Chanvorachote, P.; Muenzner, J.K.; Huebner, K.; Steinmann, S.; Erlenbach-Wuensch, K.; Geppert, C.I.; Agaimy, A.; et al. Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal cancer cells. Cell Death Dis. 2019, 10, 379. [Google Scholar] [CrossRef] [Green Version]
  38. Lang, M.; Borgmann, M.; Oberhuber, G.; Evstatiev, R.; Jimenez, K.; Dammann, K.W.; Jambrich, M.; Khare, V.; Campregher, C.; Ristl, R.; et al. Thymoquinone attenuates tumor growth in ApcMin mice by interference with Wnt-signaling. Mol. Cancer 2013, 12, 41. [Google Scholar] [CrossRef] [Green Version]
  39. Ma, J.; Zhang, Y.; Deng, H.; Liu, Y.; Lei, X.; He, P.; Dong, W. Thymoquinone inhibits the proliferation and invasion of esophageal cancer cells by disrupting the AKT/GSK-3β/Wnt signaling pathway via PTEN upregulation. Phytother. Res. 2020, 34, 3388–3399. [Google Scholar] [CrossRef]
  40. Ibrahim, A.; Alhosin, M.; Papin, C.; Ouararhni, K.; Omran, Z.; Zamzami, M.A.; Al-Malki, A.L.; Choudhry, H.; Mély, Y.; Hamiche, A.; et al. Thymoquinone challenges UHRF1 to commit auto-ubiquitination: A key event for apoptosis induction in cancer cells. Oncotarget 2018, 9, 28599–28611. [Google Scholar] [CrossRef]
  41. Kim, M.J.; Lee, H.J.; Choi, M.Y.; Kang, S.S.; Kim, Y.S.; Shin, J.K.; Choi, W.S. UHRF1 induces methylation of the TXNIP promoter and down-regulates gene expression in cervical cancer. Mol. Cells 2021, 44, 146–159. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, H.J.; Kim, M.J.; Kim, Y.S.; Choi, M.Y.; Cho, G.J.; Choi, W.S. UHRF1 silences gelsolin to inhibit cell death in early stage cervical cancer. Biochem. Biophys. Res. Commun. 2020, 526, 1061–1068. [Google Scholar] [CrossRef] [PubMed]
  43. Hennessy, B.T.; Smith, D.L.; Ram, P.T.; Lu, Y.; Mills, G.B. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug Discov. 2005, 4, 988–1004. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Janku, F.; Yap, T.A.; Meric-Bernstam, F. Targeting the PI3K pathway in cancer: Are we making headway? Nat. Rev. Clin. Oncol. 2018, 15, 273–291. [Google Scholar] [CrossRef] [PubMed]
  46. Janku, F.; Hong, D.S.; Fu, S.; Piha-Paul, S.A.; Naing, A.; Falchook, G.S.; Tsimberidou, A.M.; Stepanek, V.M.; Moulder, S.L.; Lee, J.J.; et al. Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors. Cell Rep. 2014, 6, 377–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Ihle, N.T.; Lemos, R., Jr.; Wipf, P.; Yacoub, A.; Mitchell, C.; Siwak, D.; Mills, G.B.; Dent, P.; Kirkpatrick, D.L.; Powis, G. Mutations in the phosphatidylinositol-3-kinase pathway predict for antitumor activity of the inhibitor PX-866 whereas oncogenic Ras is a dominant predictor for resistance. Cancer Res. 2009, 69, 143–150. [Google Scholar] [CrossRef] [Green Version]
  48. Feng, L.M.; Wang, X.F.; Huang, Q.X. Thymoquinone induces cytotoxicity and reprogramming of EMT in gastric cancer cells by targeting PI3K/Akt/mTOR pathway. J. Biosci. 2017, 42, 547–554. [Google Scholar] [CrossRef]
  49. Mu, G.G.; Zhang, L.L.; Li, H.Y.; Liao, Y.; Yu, H.G. Thymoquinone pretreatment overcomes the insensitivity and potentiates the antitumor effect of gemcitabine through abrogation of Notch1, PI3K/Akt/mTOR regulated signaling pathways in pancreatic cancer. Dig. Dis. Sci. 2015, 60, 1067–1080. [Google Scholar] [CrossRef]
  50. Zhang, Y.; Fan, Y.; Huang, S.; Wang, G.; Han, R.; Lei, F.; Luo, A.; Jing, X.; Zhao, L.; Gu, S.; et al. Thymoquinone inhibits the metastasis of renal cell cancer cells by inducing autophagy via AMPK/mTOR signaling pathway. Cancer Sci. 2018, 109, 3865–3873. [Google Scholar] [CrossRef]
  51. Idris, S.; Refaat, B.; Almaimani, R.A.; Ahmed, H.G.; Ahmad, J.; Alhadrami, M.; El-Readi, M.Z.; Elzubier, M.E.; Alaufi, H.A.A.; Al-Amin, B.; et al. Enhanced in vitro tumoricidal effects of 5-Fluorouracil, thymoquinone, and active vitamin D3 triple therapy against colon cancer cells by attenuating the PI3K/AKT/mTOR pathway. Life Sci. 2022, 296, 120442. [Google Scholar] [CrossRef] [PubMed]
  52. Pikarsky, E.; Porat, R.M.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar] [CrossRef] [PubMed]
  53. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Senftleben, U.; Cao, Y.; Xiao, G.; Greten, F.R.; Krähn, G.; Bonizzi, G.; Chen, Y.; Hu, Y.; Fong, A.; Sun, S.C.; et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 2001, 293, 1495–1499. [Google Scholar] [CrossRef]
  55. Erez, N.; Truitt, M.; Olson, P.; Arron, S.T.; Hanahan, D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef] [Green Version]
  56. Kabil, N.; Bayraktar, R.; Kahraman, N.; Mokhlis, H.A.; Calin, G.A.; Lopez-Berestein, G.; Ozpolat, B. Thymoquinone inhibits cell proliferation, migration, and invasion by regulating the elongation factor 2 kinase (eEF-2K) signaling axis in triple-negative breast cancer. Breast Cancer Res. Treat. 2018, 171, 593–605. [Google Scholar] [CrossRef]
  57. Siveen, K.S.; Mustafa, N.; Li, F.; Kannaiyan, R.; Ahn, K.S.; Kumar, A.P.; Chng, W.J.; Sethi, G. Thymoquinone overcomes chemoresistance and enhances the anticancer effects of bortezomib through abrogation of NF-κB regulated gene products in multiple myeloma xenograft mouse model. Oncotarget 2014, 5, 634–648. [Google Scholar] [CrossRef] [Green Version]
  58. Jafri, S.H.; Glass, J.; Shi, R.; Zhang, S.; Prince, M.; Kleiner-Hancock, H. Thymoquinone and cisplatin as a therapeutic combination in lung cancer: In vitro and in vivo. J. Exp. Clin. Cancer Res. 2010, 29, 87. [Google Scholar] [CrossRef] [Green Version]
  59. Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Chen, R.J.; Lin, Y.M.; Viswanadha, V.P.; Kuo, W.W.; Huang, C.Y. Inhibition of NF-κB and metastasis in irinotecan (CPT-11)-resistant LoVo colon cancer cells by thymoquinone via JNK and p38. Environ. Toxicol. 2017, 32, 669–678. [Google Scholar] [CrossRef]
  60. Alshyarba, M.; Otifi, H.; Al Fayi, M.; ADera, A.; Rajagopalan, P. Thymoquinone inhibits IL-7-induced tumor progression and metastatic invasion in prostate cancer cells by attenuating matrix metalloproteinase activity and Akt/NF-κB signaling. Biotechnol. Appl. Biochem. 2021, 68, 1403–1411. [Google Scholar] [CrossRef]
  61. Wilson, A.J.; Saskowski, J.; Barham, W.; Yull, F.; Khabele, D. Thymoquinone enhances cisplatin-response through direct tumor effects in a syngeneic mouse model of ovarian cancer. J. Ovarian Res. 2015, 8, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Wilson, A.J.; Saskowski, J.; Barham, W.; Khabele, D.; Yull, F. Microenvironmental effects limit efficacy of thymoquinone treatment in a mouse model of ovarian cancer. Mol. Cancer 2015, 14, 192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Shanmugam, M.K.; Ahn, K.S.; Hsu, A.; Woo, C.C.; Yuan, Y.; Tan, K.H.B.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Koh, A.P.F.; et al. Thymoquinone inhibits bone metastasis of breast cancer cells through abrogation of the CXCR4 signaling axis. Front. Pharmacol. 2018, 9, 1294. [Google Scholar] [CrossRef] [PubMed]
  64. Badr, G.; Lefevre, E.A.; Mohany, M. Thymoquinone inhibits the CXCL12-induced chemotaxis of multiple myeloma cells and increases their susceptibility to Fas-mediated apoptosis. PLoS ONE 2011, 6, e23741. [Google Scholar] [CrossRef]
  65. Pitti, R.M.; Marsters, S.A.; Ruppert, S.; Donahue, C.J.; Moore, A.; Ashkenazi, A. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J. Biol. Chem. 1996, 271, 12687–12690. [Google Scholar] [CrossRef] [Green Version]
  66. Walczak, H.; Miller, R.E.; Ariail, K.; Gliniak, B.; Griffith, T.S.; Kubin, M.; Chin, W.; Jones, J.; Woodward, A.; Le, T.; et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat. Med. 1999, 5, 157–163. [Google Scholar] [CrossRef]
  67. Pan, G.; O’Rourke, K.; Chinnaiyan, A.M.; Gentz, R.; Ebner, R.; Ni, J.; Dixit, V.M. The receptor for the cytotoxic ligand TRAIL. Science 1997, 276, 111–113. [Google Scholar] [CrossRef]
  68. Zhang, X.D.; Franco, A.; Myers, K.; Gray, C.; Nguyen, T.; Hersey, P. Relation of TNF-related apoptosis-inducing ligand (TRAIL) receptor and FLICE-inhibitory protein expression to TRAIL-induced apoptosis of melanoma. Cancer Res. 1999, 59, 2747–2753. [Google Scholar]
  69. Gliniak, B.; Le, T. Tumor necrosis factor-related apoptosis-inducing ligand’s antitumor activity in vivo is enhanced by the chemotherapeutic agent CPT-11. Cancer Res. 1999, 59, 6153–6158. [Google Scholar]
  70. Roth, W.; Isenmann, S.; Naumann, U.; Kügler, S.; Bähr, M.; Dichgans, J.; Ashkenazi, A.; Weller, M. Locoregional Apo2L/TRAIL eradicates intracranial human malignant glioma xenografts in athymic mice in the absence of neurotoxicity. Biochem. Biophys. Res. Commun. 1999, 265, 479–483. [Google Scholar] [CrossRef]
  71. Zhang, R.; Wu, T.; Zheng, P.; Liu, M.; Xu, G.; Xi, M.; Yu, J. Thymoquinone sensitizes human hepatocarcinoma cells to TRAIL-induced apoptosis via oxidative DNA damage. DNA Repair. 2021, 103, 103117. [Google Scholar] [CrossRef] [PubMed]
  72. Ashour, A.E.; Abd-Allah, A.R.; Korashy, H.M.; Attia, S.M.; Alzahrani, A.Z.; Saquib, Q.; Bakheet, S.A.; Abdel-Hamied, H.E.; Jamal, S.; Rishi, A.K. Thymoquinone suppression of the human hepatocellular carcinoma cell growth involves inhibition of IL-8 expression, elevated levels of TRAIL receptors, oxidative stress and apoptosis. Mol. Cell. Biochem. 2014, 389, 85–98. [Google Scholar] [CrossRef] [PubMed]
  73. Ashour, A.E.; Ahmed, A.F.; Kumar, A.; Zoheir, K.M.; Aboul-Soud, M.A.; Ahmad, S.F.; Attia, S.M.; Abd-Allah, A.R.; Cheryan, V.T.; Rishi, A.K. Thymoquinone inhibits growth of human medulloblastoma cells by inducing oxidative stress and caspase-dependent apoptosis while suppressing NF-κB signaling and IL-8 expression. Mol. Cell. Biochem. 2016, 416, 141–155. [Google Scholar] [CrossRef] [PubMed]
  74. Hussain, A.R.; Uddin, S.; Ahmed, M.; Al-Dayel, F.; Bavi, P.P.; Al-Kuraya, K.S. Phosphorylated IκBα predicts poor prognosis in activated B-cell lymphoma and its inhibition with thymoquinone induces apoptosis via ROS release. PLoS ONE 2013, 8, e60540. [Google Scholar] [CrossRef] [Green Version]
  75. Hussain, A.R.; Ahmed, M.; Ahmed, S.; Manogaran, P.; Platanias, L.C.; Alvi, S.N.; Al-Kuraya, K.S.; Uddin, S. Thymoquinone suppresses growth and induces apoptosis via generation of reactive oxygen species in primary effusion lymphoma. Free Radic. Biol. Med. 2011, 50, 978–987. [Google Scholar] [CrossRef]
  76. Brannon, A.R.; Vakiani, E.; Sylvester, B.E.; Scott, S.N.; McDermott, G.; Shah, R.H.; Kania, K.; Viale, A.; Oschwald, D.M.; Vacic, V.; et al. Comparative sequencing analysis reveals high genomic concordance between matched primary and metastatic colorectal cancer lesions. Genome Biol. 2014, 15, 454. [Google Scholar] [CrossRef]
  77. Goswami, R.S.; Patel, K.P.; Singh, R.R.; Meric-Bernstam, F.; Kopetz, E.S.; Subbiah, V.; Alvarez, R.H.; Davies, M.A.; Jabbar, K.J.; Roy-Chowdhuri, S.; et al. Hotspot mutation panel testing reveals clonal evolution in a study of 265 paired primary and metastatic tumors. Clin. Cancer Res. 2015, 21, 2644–2651. [Google Scholar] [CrossRef] [Green Version]
  78. Vignot, S.; Lefebvre, C.; Frampton, G.M.; Meurice, G.; Yelensky, R.; Palmer, G.; Capron, F.; Lazar, V.; Hannoun, L.; Miller, V.A.; et al. Comparative analysis of primary tumour and matched metastases in colorectal cancer patients: Evaluation of concordance between genomic and transcriptional profiles. Eur. J. Cancer 2015, 51, 791–799. [Google Scholar] [CrossRef]
  79. Johnson, B.E.; Mazor, T.; Hong, C.; Barnes, M.; Aihara, K.; McLean, C.Y.; Fouse, S.D.; Yamamoto, S.; Ueda, H.; Tatsuno, K.; et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 2014, 343, 189–193. [Google Scholar] [CrossRef] [Green Version]
  80. Murugaesu, N.; Wilson, G.A.; Birkbak, N.J.; Watkins, T.; McGranahan, N.; Kumar, S.; Abbassi-Ghadi, N.; Salm, M.; Mitter, R.; Horswell, S.; et al. Tracking the genomic evolution of esophageal adenocarcinoma through neoadjuvant chemotherapy. Cancer Discov. 2015, 5, 821–831. [Google Scholar] [CrossRef] [Green Version]
  81. Iskender, B.; Izgi, K.; Canatan, H. Novel anti-cancer agent myrtucommulone-A and thymoquinone abrogate epithelial-mesenchymal transition in cancer cells mainly through the inhibition of PI3K/AKT signalling axis. Mol. Cell. Biochem. 2016, 416, 71–84. [Google Scholar] [CrossRef] [PubMed]
  82. Li, J.; Khan, M.A.; Wei, C.; Cheng, J.; Chen, H.; Yang, L.; Ijaz, I.; Fu, J. Thymoquinone inhibits the migration and invasive characteristics of cervical cancer cells SiHa and CaSki in vitro by targeting epithelial to mesenchymal transition associated transcription factors Twist1 and Zeb1. Molecules 2017, 22, 2105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jazirehi, A.R.; Vega, M.I.; Chatterjee, D.; Goodglick, L.; Bonavida, B. Inhibition of the Raf-MEK1/2-ERK1/2 signaling pathway, Bcl-xL down-regulation, and chemosensitization of non-Hodgkin’s lymphoma B cells by Rituximab. Cancer Res. 2004, 64, 7117–7126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yeung, K.; Seitz, T.; Li, S.; Janosch, P.; McFerran, B.; Kaiser, C.; Fee, F.; Katsanakis, K.D.; Rose, D.W.; Mischak, H.; et al. Suppression of Raf-1 kinase activity and MAP kinase signalling by RKIP. Nature 1999, 401, 173–177. [Google Scholar] [CrossRef] [Green Version]
  85. Schoeberl, B.; Eichler-Jonsson, C.; Gilles, E.D.; Müller, G. Computational modeling of the dynamics of the MAP kinase cascade activated by surface and internalized EGF receptors. Nat. Biotechnol. 2002, 20, 370–375. [Google Scholar] [CrossRef]
  86. Teis, D.; Wunderlich, W.; Huber, L.A. Localization of the MP1-MAPK scaffold complex to endosomes is mediated by p14 and required for signal transduction. Dev. Cell 2002, 3, 803–814. [Google Scholar] [CrossRef] [Green Version]
  87. Schaeffer, H.J.; Catling, A.D.; Eblen, S.T.; Collier, L.S.; Krauss, A.; Weber, M.J. MP1: A MEK binding partner that enhances enzymatic activation of the MAP kinase cascade. Science 1998, 281, 1668–1671. [Google Scholar] [CrossRef]
  88. Troppmair, J.; Bruder, J.T.; Munoz, H.; Lloyd, P.A.; Kyriakis, J.; Banerjee, P.; Avruch, J.; Rapp, U.R. Mitogen-activated protein kinase/extracellular signal-regulated protein kinase activation by oncogenes, serum, and 12-O-tetradecanoylphorbol-13-acetate requires Raf and is necessary for transformation. J. Biol. Chem. 1994, 269, 7030–7035. [Google Scholar] [CrossRef]
  89. Sebolt-Leopold, J.S. Development of anticancer drugs targeting the MAP kinase pathway. Oncogene 2000, 19, 6594–6599. [Google Scholar] [CrossRef] [Green Version]
  90. Smalley, K.S. A pivotal role for ERK in the oncogenic behaviour of malignant melanoma? Int. J. Cancer 2003, 104, 527–532. [Google Scholar] [CrossRef]
  91. Woo, C.C.; Hsu, A.; Kumar, A.P.; Sethi, G.; Tan, K.H. Thymoquinone inhibits tumor growth and induces apoptosis in a breast cancer xenograft mouse model: The role of p38 MAPK and ROS. PLoS ONE 2013, 8, e75356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. El-Baba, C.; Mahadevan, V.; Fahlbusch, F.B.; Mohan, S.S.; Rau, T.T.; Gali-Muhtasib, H.; Schneider-Stock, R. Thymoquinone-induced conformational changes of PAK1 interrupt prosurvival MEK-ERK signaling in colorectal cancer. Mol. Cancer 2014, 13, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Dirican, A.; Atmaca, H.; Bozkurt, E.; Erten, C.; Karaca, B.; Uslu, R. Novel combination of docetaxel and thymoquinone induces synergistic cytotoxicity and apoptosis in DU-145 human prostate cancer cells by modulating PI3K-AKT pathway. Clin. Transl. Oncol. 2015, 17, 145–151. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, J.; Kuang, X.R.; Lv, P.T.; Yan, X.X. Thymoquinone inhibits proliferation and invasion of human nonsmall-cell lung cancer cells via ERK pathway. Tumour Biol. 2015, 36, 259–269. [Google Scholar] [CrossRef] [PubMed]
  95. El-Najjar, N.; Chatila, M.; Moukadem, H.; Vuorela, H.; Ocker, M.; Gandesiri, M.; Schneider-Stock, R.; Gali-Muhtasib, H. Reactive oxygen species mediate thymoquinone-induced apoptosis and activate ERK and JNK signaling. Apoptosis 2010, 15, 183–195. [Google Scholar] [CrossRef] [PubMed]
  96. Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Lytle, J.R.; Yario, T.A.; Steitz, J.A. Target mRNAs are repressed as efficiently by microRNA-binding sites in the 5’ UTR as in the 3’ UTR. Proc. Natl. Acad. Sci. USA 2007, 104, 9667–9672. [Google Scholar] [CrossRef] [Green Version]
  98. Khraiwesh, B.; Arif, M.A.; Seumel, G.I.; Ossowski, S.; Weigel, D.; Reski, R.; Frank, W. Transcriptional control of gene expression by microRNAs. Cell 2010, 140, 111–122. [Google Scholar] [CrossRef]
  99. Zhou, X.; Wang, F.; Wu, H.; Chen, X.; Zhang, Y.; Lin, J.; Cai, Y.; Xiang, J.; He, N.; Hu, Z.; et al. Thymoquinone suppresses the proliferation, migration and invasiveness through regulating ROS, autophagic flux and miR-877-5p in human bladder carcinoma cells. Int. J. Biol. Sci. 2021, 17, 3456–3475. [Google Scholar] [CrossRef]
  100. Bashir, A.O.; El-Mesery, M.E.; Anwer, R.; Eissa, L.A. Thymoquinone potentiates miR-16 and miR-375 expressions in hepatocellular carcinoma. Life Sci. 2020, 254, 117794. [Google Scholar] [CrossRef]
  101. Bhattacharya, S.; Ghosh, A.; Maiti, S.; Ahir, M.; Debnath, G.H.; Gupta, P.; Bhattacharjee, M.; Ghosh, S.; Chattopadhyay, S.; Mukherjee, P.; et al. Delivery of thymoquinone through hyaluronic acid-decorated mixed Pluronic® nanoparticles to attenuate angiogenesis and metastasis of triple-negative breast cancer. J. Control. Release 2020, 322, 357–374. [Google Scholar] [CrossRef] [PubMed]
  102. Upadhyay, P.; Sarker, S.; Ghosh, A.; Gupta, P.; Das, S.; Ahir, M.; Bhattacharya, S.; Chattopadhyay, S.; Ghosh, S.; Adhikary, A. Transferrin-decorated thymoquinone-loaded PEG-PLGA nanoparticles exhibit anticarcinogenic effect in non-small cell lung carcinoma via the modulation of miR-34a and miR-16. Biomater. Sci. 2019, 7, 4325–4344. [Google Scholar] [CrossRef] [PubMed]
  103. Bhattacharya, S.; Ahir, M.; Patra, P.; Mukherjee, S.; Ghosh, S.; Mazumdar, M.; Chattopadhyay, S.; Das, T.; Chattopadhyay, D.; Adhikary, A. PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a. Biomaterials 2015, 51, 91–107. [Google Scholar] [CrossRef]
  104. Wei, C.; Zou, H.; Xiao, T.; Liu, X.; Wang, Q.; Cheng, J.; Fu, S.; Peng, J.; Xie, X.; Fu, J. TQFL12, a novel synthetic derivative of TQ, inhibits triple-negative breast cancer metastasis and invasion through activating AMPK/ACC pathway. J. Cell Mol. Med. 2021, 25, 10101–10110. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, S.R.; Mun, J.Y.; Jeong, M.S.; Lee, H.H.; Roh, Y.G.; Kim, W.T.; Kim, M.H.; Heo, J.; Choi, Y.H.; Kim, S.J.; et al. Thymoquinone-induced tristetraprolin inhibits tumor growth and metastasis through destabilization of MUC4 mRNA. Int. J Mol. Sci. 2019, 20, 2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Dera, A.A.; Rajagopalan, P.; Al Fayi, M.; Ahmed, I.; Chandramoorthy, H.C. Indirubin-3-monoxime and thymoquinone exhibit synergistic efficacy as therapeutic combination in in-vitro and in-vivo models of Lung cancer. Arch. Pharm. Res. 2020, 43, 655–665. [Google Scholar] [CrossRef] [PubMed]
  107. Yi, T.; Cho, S.G.; Yi, Z.; Pang, X.; Rodriguez, M.; Wang, Y.; Sethi, G.; Aggarwal, B.B.; Liu, M. Thymoquinone inhibits tumor angiogenesis and tumor growth through suppressing AKT and extracellular signal-regulated kinase signaling pathways. Mol. Cancer Ther. 2008, 7, 1789–1796. [Google Scholar] [CrossRef] [Green Version]
  108. Ahmad, I.; Muneer, K.M.; Tamimi, I.A.; Chang, M.E.; Ata, M.O.; Yusuf, N. Thymoquinone suppresses metastasis of melanoma cells by inhibition of NLRP3 inflammasome. Toxicol. Appl. Pharmacol. 2013, 270, 70–76. [Google Scholar] [CrossRef]
  109. Singh, S.K.; Gordetsky, J.B.; Bae, S.; Acosta, E.P.; Lillard, J.W., Jr.; Singh, R. Selective targeting of the hedgehog signaling pathway by PBM nanoparticles in docetaxel-resistant prostate cancer. Cells 2020, 9, 1976. [Google Scholar] [CrossRef]
  110. Kou, B.; Liu, W.; Zhao, W.; Duan, P.; Yang, Y.; Yi, Q.; Guo, F.; Li, J.; Zhou, J.; Kou, Q. Thymoquinone inhibits epithelial-mesenchymal transition in prostate cancer cells by negatively regulating the TGF-β/Smad2/3 signaling pathway. Oncol. Rep. 2017, 38, 3592–3598. [Google Scholar] [CrossRef] [Green Version]
  111. Kensara, O.A.; El-Shemi, A.G.; Mohamed, A.M.; Refaat, B.; Idris, S.; Ahmad, J. Thymoquinone subdues tumor growth and potentiates the chemopreventive effect of 5-fluorouracil on the early stages of colorectal carcinogenesis in rats. Drug Des. Devel. Ther. 2016, 10, 2239–2253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Mohamed, A.M.; Refaat, B.A.; El-Shemi, A.G.; Kensara, O.A.; Ahmad, J.; Idris, S. Thymoquinone potentiates chemoprotective effect of Vitamin D3 against colon cancer: A pre-clinical finding. Am. J. Transl. Res. 2017, 9, 774–790. [Google Scholar] [PubMed]
Figure 1. (A) JAK/STAT-mediated signaling is regulated by thymoquinone. (B,C) Thymoquinone inhibited JAK2 and STAT3. Thymoquinone stimulated the expression of SOCS1 and SOCS3. (D) STAT3-mediated upregulation of VEGF; BCL-2; and cyclin D1, D2, and D3.
Figure 1. (A) JAK/STAT-mediated signaling is regulated by thymoquinone. (B,C) Thymoquinone inhibited JAK2 and STAT3. Thymoquinone stimulated the expression of SOCS1 and SOCS3. (D) STAT3-mediated upregulation of VEGF; BCL-2; and cyclin D1, D2, and D3.
Ijms 23 06311 g001
Figure 2. Metastatic spread of cancer cells to distant organs.
Figure 2. Metastatic spread of cancer cells to distant organs.
Ijms 23 06311 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Farooqi, A.A.; Attar, R.; Xu, B. Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone. Int. J. Mol. Sci. 2022, 23, 6311. https://doi.org/10.3390/ijms23116311

AMA Style

Farooqi AA, Attar R, Xu B. Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone. International Journal of Molecular Sciences. 2022; 23(11):6311. https://doi.org/10.3390/ijms23116311

Chicago/Turabian Style

Farooqi, Ammad Ahmad, Rukset Attar, and Baojun Xu. 2022. "Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone" International Journal of Molecular Sciences 23, no. 11: 6311. https://doi.org/10.3390/ijms23116311

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop