Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
November-2025 Volume 30 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
November-2025 Volume 30 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Article Open Access

Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity

  • Authors:
    • Nouf A. Aldarmahi
    • Nesrin I. Tarbiah
    • Nuha A. Alkhattabi
    • Huda F. Alshaibi
  • View Affiliations / Copyright

    Affiliations: Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
    Copyright: © Aldarmahi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 530
    |
    Published online on: September 18, 2025
       https://doi.org/10.3892/ol.2025.15276
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Dark cumin, or Nigella sativa, is a well‑known and popular herbal medicine that may be used in the treatment of gastrointestinal issues, diabetes, bronchitis, high blood pressure and cancer. The anticancer action of Nigella sativa may be associated with the immune‑modulatory effect of its activation of human natural killer (NK) cells. NK cells, a subset of lymphocytes, are a component of innate immunity, the body's initial line of defense against infections. To identify potential ways to improve immunotherapy using natural products, the present study aimed to assess the impact of thymoquinone (TQ), a key component of Nigella sativa, on the activity of NK cells through their cytotoxicity and released chemicals. CD56 NK cells that were isolated and positively selected from blood samples, were cultured with PC3‑RFP prostate cancer cells and treated with TQ at different concentrations (25 and 50 µM). Cytotoxicity was measured, and perforin, granzyme B and interferon‑α (IFN‑α) levels were assessed using ELISA. The results demonstrated that the cytotoxic effect of NK cells on PC3‑RFP was significantly increased in the presence of TQ from 55.1 to 85.35%. Furthermore, with 50 µM TQ, there was a significant increase in secretion of perforin (0.80 ng/ml) compared with the control (0.2 ng/ml), and of granzyme B (1.24 ng/ml) compared with the control (0.65 ng/ml). Moreover, the production of IFN‑α by the NK cells significantly increased in the presence of 25 and 50 µM TQ (0.5 and 0.7 ng/ml, respectively) compared with the control (0.3 ng/ml). In summary, TQ increased NK cell cytotoxicity against PC3‑RFP cells, indicating a potential application in the development of cell‑based immunity treatment that requires more research and has promising prospects. 

Introduction

Globally, the burden of cancer incidence and death is increasing rapidly (1). Cancer is a complex disease and there is no single treatment that is effective for all types (2,3). With an estimated 1,414,259 new cases worldwide each year, prostate cancer (PC) is the second most frequent malignancy and the top cause of death for men worldwide (4). The average age at diagnosis is 66 years old, and PC incidence and mortality rise with age globally (3). There are 158.3 new cases diagnosed per 100,000 males, with 293,818 additional cases projected by 2040 (5). Moreover, African-American men have been reported to experience higher incidence rates and roughly double the mortality rate from PC than Caucasian men. This discrepancy has been explained by social, environmental and genetic variations (2).

PC that is detected early may not show any symptoms, often advances slowly and may be treated with little-to-no intervention. The most frequent symptoms, however, are nocturia, increased frequency of urine and difficulty urinating, which are indicative of prostatic enlargement. In more advanced stages of the disease, patients may experience back discomfort and urine retention due to metastasis to the axis skeleton, the most common site of bone metastatic disease (6).

The risk of PC is notably influenced by diet and exercise. For example, dietary factors are associated with the observed variations in PC incidence rates between countries and ethnic groups (7,8). However, the majority of studies concentrate on the genes implicated in the inherited form of PC as well as the mutations that take place in the acquired form (6,7). Therefore, a thorough analysis of PC epidemiology and risk factors may aid in understanding the relationship between genetic abnormalities and the function of the environment in causing these alterations and/or encouraging tumor progression. These differences in PC incidence imply that the etiology of PC is markedly influenced by environmental variables. However, underdiagnosis, disparities in screening techniques and access to healthcare may also contribute to variations in incidence (6).

Due to several biological and tumor microenvironmental factors, PC presents substantial treatment challenges. For example, prostate tumors often have a low tumor mutational burden, meaning that the immune system targets fewer neoantigens (9). Furthermore, immunosuppressive cells, such regulatory T cells, tumor-associated macrophages and myeloid-derived suppressor cells, are present in the PC immunosuppressive tumor microenvironment. The normal immune response against cancer cells is hampered by these cells, which produce a ‘cold’ tumor environment with limited immune cell penetration (10). Furthermore, although androgen deprivation therapy is initially successful, certain patients experience resistance to the treatment, which can result in metastatic castration-resistant PC. Changes in the tumor microenvironment and immune evasion strategies are associated with this development (11).

Using immune checkpoint inhibitors, cancer vaccines, chimeric antigen receptor T-cell treatment and combination medicines, immunotherapy offers potential treatments by using the immune system to target and kill cancer cells. By reducing antitumor immune responses or modifying tumor immunogenicity, several immune system components cooperate to either protect the host against necessary tumor progression, enhance tumor escape, or both (12). This process is known as cancer immuno-altering (12,13). Immune spots, which are resistant cell surface receptors that regulate the suppression or activation of immune reactions, serve an important role in different phases of the immune responses. For example, both PD-1 (Programmed Death-1) and cytotoxic T-Lymphocyte-Associated Protein 4 CTLA4, prevent overactivation of the immune system once binding to their ligands (14). The optimum outcome for controlling tumors is immune system activation (13). In addition to surgery, chemotherapy, radiation and targeted medicines, cancer immunotherapy has become a crucial therapeutic approach. Furthermore, cancer immunotherapy has demonstrated encouraging outcomes with several cancer types and can be used in conjunction with other treatments. Consequently, immunotherapy has been reported to have notable benefits: In one study (15) immunotherapy improved lung cancer treatment PD-1 inhibitor to standard chemotherapy, increased objective response rate and progression free survival. In addition, another study showed that combining immunotherapy with radiation enhance anti-tumor immune response through increasing the release of tumor antigen following radiation (16).

A subset of lymphocytes, known as natural killer (NK) cells, is essential to the innate immune response against malignancies and infections (17,18). NK cells are able to recognize and target cancerous and contaminated cells (19,20), and ~90% of the circulating NK cell population is made up of CD56 and CD16 cells, which cause cell lysis by releasing cytolytic granules that include granzyme and perforin. Together, NK cells and interferon-γ (IFN-γ) cause target cells to undergo apoptosis (21,22). For aberrant cells, NK cells increase antibody-mediated cytotoxicity and cytotoxic T lymphocytes act in a similar manner (23).

NK cells help to defend the body against foreign (pathogens) and endogenous (cancer) threats. Extensive research has been performed with the goal of identifying an efficient method for targeting impacted cells by utilizing NK cell pathways (24) (Fig. 1). However, notwithstanding the promising results of this research, tumor cells develop a defense mechanism that thwarts the cytotoxicity caused by NK cells (25,26).

Cytotoxicity mechanism of NK cells.
By evaluating the net input of activating and inhibitory signals
from target cells through NK cell surface receptors, NK cells
discriminate healthy cells from target cells. However, several
tumors develop mechanisms to escape NK cell immune responses by
modifying their cell surface molecules, such as low immunogenicity.
Following the identification of the target cell by NK cells, the
stimulation domain is activated. This is initiated by the secretion
of a cytolytic enzyme, known as perforin, stored in NK cell
vesicles. Perforin creates pores in the cell membrane of the target
cell, allowing the passage of granzymes into the target cell and
starting protein digestion (cytolysis). NK cells express TRAIL and
FasL, which interact with TRAIL receptors and FAS ligands in cancer
cells, respectively. This interaction results in cell death,
signaling complex stimulating apoptosis. NK, natural killer; TRAIL,
TNF-related apoptosis inducing ligand; TRAILR, TRAIL receptor; MHC,
major histocompatibility complex; FasL, FAS ligand. Created in
BioRender.com.

Figure 1.

Cytotoxicity mechanism of NK cells. By evaluating the net input of activating and inhibitory signals from target cells through NK cell surface receptors, NK cells discriminate healthy cells from target cells. However, several tumors develop mechanisms to escape NK cell immune responses by modifying their cell surface molecules, such as low immunogenicity. Following the identification of the target cell by NK cells, the stimulation domain is activated. This is initiated by the secretion of a cytolytic enzyme, known as perforin, stored in NK cell vesicles. Perforin creates pores in the cell membrane of the target cell, allowing the passage of granzymes into the target cell and starting protein digestion (cytolysis). NK cells express TRAIL and FasL, which interact with TRAIL receptors and FAS ligands in cancer cells, respectively. This interaction results in cell death, signaling complex stimulating apoptosis. NK, natural killer; TRAIL, TNF-related apoptosis inducing ligand; TRAILR, TRAIL receptor; MHC, major histocompatibility complex; FasL, FAS ligand. Created in BioRender.com.

Research is investigating the use of NK cells to treat (27–29) PC. NK cells can directly target and eliminate PC cells, resulting in direct tumor cytotoxicity. Moreover, research has reported that, both in vitro and in vivo, genetically modified NK cells, such as prostate-specific membrane antigen (PSMA)-targeted chimeric antigen receptor-NK cells, have strong cytotoxic effects on PC cells. They can specifically identify and eliminate PSMA-expressing PC cells, providing a targeted therapy strategy. Therefore, these altered NK cells may be employed as an ‘off-the-shelf’ therapeutic option, enabling a sustainable and economical treatment technique (27).

Additionally, PC cells usually develop defenses against immune detection, such as by producing soluble NK cell receptor D (NKG2D) ligands to obstruct the recognition of NK cells. However, NK cells can adapt to these challenges, and strategies are being developed to make them more effective against these tumor escape tactics (28). Investigating NK cell function against PC is challenging as they produce TGF-β, IL-10 and regulatory T cells, inhibiting NK activation, and have low infiltration of NK cells making it hard to study their direct interaction with PC. PC is a useful model for researching NK cell-mediated therapy, for example due to the immune microenvironment dynamics, in which PC tumors are distinguished by a distinct immune milieu that consists of both immuno-suppressive and immune-activating components (29–32). As studies have reported that NK cells in patients with PC frequently have compromised cytotoxic function, this environment offers a realistic setting for researching how NK cells interact with tumor cells and the factors determining their efficacy (29,33). This malfunction provides a clear target for therapeutic intervention trials targeted at restoring NK cell activity and is associated with the course of the disease. Furthermore, research employing preclinical models has reported that NK cells are capable of efficiently inhibiting the proliferation of PC cells resistant to castration. These results highlight how effective NK cell-based treatments can be in treating advanced PC (30). Nevertheless, PC uses several strategies to avoid immune monitoring, especially from NK cells. These tactics include modulating the expression of NK cell receptors, secreting immunosuppressive substances and changing the expression of ligands (28). PC cells can decrease the expression of ligands such NKG2D and DNAX accessory molecule 1 that are recognized by NK cell receptors. Moreover, immunosuppressive cytokines such as TGF-β are abundant in the tumor microenvironment. The release of TGF-β causes the activating receptors of NK cells, including NK cell P30-related protein and NKG2D, to be downregulated, which reduces the cytotoxic effects (31). Furthermore, PC can cause NK cells to exhibit a fatigued phenotype, which is typified by decreased activating receptor expression and decreased cytotoxic activity. This fatigue is associated with NK cells expressing more inhibitory receptors, such as T cell immunoglobulin mucin 3 and programmed cell death protein 1 (PD-1), which results in weakened antitumor responses (32). Additionally, programmed death-ligand 1 (PD-L1) expression on cancer cells may be upregulated in PC with hypoxic circumstances. Immune evasion is facilitated by the inhibition of the cytotoxic function of NK cells caused by the interaction between PD-L1 and its receptor PD-1 (34).

Finally, due to their ability to treat numerous diseases, several plants are regarded as useful therapeutic instruments in a wide range of medical disorders. Moreover, different medicinal plants have been used worldwide to alleviate the symptoms of several illnesses for millennia (35). One well-known example with a wide range of applications is Nigella sativa, also known as dark cumin. For two millennia, populations around the world have utilized this plant, a dicotyledon of the Ranunculaceae family, as a snack, spice and nutritional supplement (36). Thymoquinone (TQ), the main active ingredient in the black seeds, has garnered interest in both traditional medicine and contemporary therapeutic research (37,38). Therefore, the present study aimed to assess how TQ, which comprises ~40% of Nigella sativa (38), affects the cytotoxic activity of NK cells against the PC3-RFP cell line. The results could aid in comprehending the potential therapeutic impact of TQ on NK cells for cell-based immunotherapy.

Materials and methods

Blood collection

Blood samples were collected at King Abdulaziz University Hospital (Jeddah, Saudi Arabia) between August and November 2021 from 10 healthy volunteers six male, four female and aged 25–45 years in EDTA tubes (BD Vacutainer) and used for NK cell isolation. All experiments were performed at King Faisal Specialist Hospital and Research Centre (Jeddah, Saudi Arabia). Inclusion criteria for enrolled participants include general good health with no chronic diseases, recent infections and immunosuppressive medication or vaccination. Exclusion criteria were smoking, pregnancy or breastfeeding, autoimmune disease and malignancy.

MACS positive isolation

NK cells were purified by positive selection of CD56 cells (CD56 MicroBeads, human, 130-050-401, Miltenyi biotec) and MACS column (130-042-201, Miltenyi biotec, USA) was used according to the manufacturer's instruction.

Cell lines and culture

Transfected PC3 cells with red fluorescence (PC3-RFP cells) with the pDSRed-monomer-Hyg-C1 plasmid to produce hygromycin-resistant cells that express RFP were provided by AlShaibi et al (39). The cells were stored at King Fahd Medical Research Center (Jeddah, Saudi Arabia) and used in the present study.

PC3-RFP cells were cultured in high-glucose DMEM (cat. no. 11965118; Thermo Fisher Scientific, Inc.), with sodium pyruvate L-glutamine and Phenol Red, and supplemented with 10% penicillin-streptomycin (10,000 U/ml; cat. no. 15140122; Thermo Fisher Scientific, Inc.). NK cells were cultured in NK MACS Medium (cat. no. 130-092-657; Miltneyi Biotec B.V. & Co. KG), supplemented with 20% FBS (cat. no. 12103C; Sigma-Aldrich; Merck KGaA) and 0.1 mM β-mercaptoethanol (Sigma-Aldrich; Merck KGaA), IL-2 (human animal-component free, recombinant, expressed in E. coli, ≥98%; cat. no. SRP3085; Sigma-Aldrich; Merck KGaA). The cells were routinely maintained in humidified incubator at 37°C and 5% CO2.

TQ preparation

TQ (cat. no. 274666-1G; Sigma-Aldrich; Merck KGaA) was prepared by dissolving 4.926 mg TQ crystal in 10 µl anhydrous dimethyl sulfoxide (≥99.9%; cat. no. 276855; Sigma-Aldrich; Merck KGaA) and 9,990 µl NK medium to yield a stock solution. The final DMSO concentration in all TQ working solutions (50 and 25 µM) was 0.1%. Vehicle controls containing 0.1% DMSO in NK medium without TQ were included in all experiments.

Cell cytotoxicity of TQ on tumor (PC3-RFP) and NK cells

Cell cytotoxicity was analyzed using the CytoTox® 96 Non-Radioactive Cytotoxicity Assay kit (Promega Corporation). Briefly, PC3-RFP was plated at a density of 15×103 cells per well in a 96-well flat-bottom plate and incubated overnight at room temperature. Subsequently, NK cells were co-cultured at an effector cell/target cell ratio of 1:2 in the presence of different concentrations (25 and 50 µM) of TQ for 5 h. CytoTox 96 lysis buffer was added and incubated for 45 min at room temperature. CytoTox 96 reagent was then added to each well and incubated for 30 min at room temperature in the dark, and then the stop solution was added. The absorbance was measured at 680 and 490 nm.

Flow cytometry

Following positive selection of NK cells were stained with CD56-PE (DAKO; Agilent Technologies, Inc.) at 25°C for 30 min in the dark. The cells were then analyzed using flow cytometry (Novocyte Flow Cytometer; Agilent Technologies, Inc.). Phycoerythrin (PE) detection channel was the analyte detector, CD56-PE was the analyte reporter, and NovoExpress software (version 1.6.3; Agilent Technologies, Inc.)used for data acquisition and analysis.

ELISA

PC-3-RFP and NK cells (1:2; 15–30×103) were cocultured in the presence of TQ for 5 h at 37°C. Cell-free supernatants were harvested and ELISA was used for detection of IFN-α (Human IFN-α ELISA Kit; cat. no. BMS216 Invitrogen™; Thermo Fisher Scientific, Inc.), granzyme B [Human granzymes B (Gzms-B) ELISA kit; cat. no. E0899Hu; Shanghai Korain Biotech Co., Ltd.] and perforin [Human Perforin/Pore-forming protein (PF/PFP) ELISA Kit; cat. no. E0070Hu; Shanghai Korain Biotech Co., Ltd.] were used. Absorbance was measured at 450 nm using a microplate reader.

Statistical analysis

The data were analyzed using GraphPad Prism 9 (IBM Corp.) and the results are presented as the mean ± standard deviation of three independent experiments. Statistical significance was assessed using one-way analysis of variance and Tukey's multiple comparison test. Spearman correlation coefficient was used for correlation tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Flow cytometry analysis

Using flow cytometry, the positivity of CD56 was determined to assess the purity of the isolated NK cells by comparison with the defined cutoff values obtained with unstained control cells. The NK cell purity was 95.7% (Fig. 2).

NK cells dot plot before treatment.
CD56 positivity was determined by comparison with defined cut-off
values obtained from unstained control cells. The NK cell purity
was 95.7%. NK, natural killer.

Figure 2.

NK cells dot plot before treatment. CD56 positivity was determined by comparison with defined cut-off values obtained from unstained control cells. The NK cell purity was 95.7%. NK, natural killer.

Cytotoxic effect of TQ on NK cells with PC3-RFP

The cytotoxicity of NK cells was evaluated in co-culture with PC3-RFP cells. The effector (NK cells) and target (PC3-RFP) ratio was 1:2 and different concentrations of TQ (25 and 50 µM) were added in the culture. In the presence of TQ, there was a significant increase in NK cell cytotoxicity: Both concentrations of TQ (25 and 50 µM) significantly upregulated NK cell cytotoxicity in PC3-RFP cells, with cytotoxicity at 85.35% compared with 55.1% in the control group of NK cells co-cultured with tumor cells. Cell cytotoxicity was also significantly increased in tumor cells treated with both 25 and 50 µM TQ in the presence of NK cells, compared with tumor cells treated with the same TQ concentrations in the absence of NK cells (Fig. 3).

NK cell cytotoxicity against PC3-RFP
cells in the presence or absence of TQ. The effector (NK cells) and
target (PC3-RFP) ratio was 1:2. *P<0.05; **P<0.01;
***P<0.001. NK, natural killer; TQ, thymoquinone; TC, tumor
cells.

Figure 3.

NK cell cytotoxicity against PC3-RFP cells in the presence or absence of TQ. The effector (NK cells) and target (PC3-RFP) ratio was 1:2. *P<0.05; **P<0.01; ***P<0.001. NK, natural killer; TQ, thymoquinone; TC, tumor cells.

Effect of TQ on the activity of NK cells

Co-culture of NK cells with PC3-RFP cells significantly increased the production of perforin, granzyme B and IFN-α from NK cells compared with NK cells alone using both concentrations of TQ (25 and 50 µM). NK cells co-cultured with PC3-RFP cells and treated with 50 µM TQ had significantly increased production of perforin and granzyme B production compared with the control NK cells co-cultured with tumor cells. The higher 50 µM TQ dose increased the production of perforin and granzyme B (0.80 and 1.24 ng/ml, respectively) by the NK cells more than the lower 25 µM dose (0.50 and 0.76 ng/ml, respectively). Furthermore, IFN-α production by the NK cells was significantly increased in the presence of 25 and 50 µM TQ (0.5 and 0.7 ng/ml, respectively) compared with the control NK cells co-cultured with tumor cells (0.3 ng/ml), with a greater increase observed in the presence of 50 µM TQ (Fig. 4).

Production of major cytokines against
PC3-RFP cells by NK cells in the presence or absence of TQ. TQ
enhances the release of (A) perforin, (B) granzyme B and (C) IFN-α,
the major secreted cytokines of NK cells. *P<0.05; **P<0.01;
***P<0.001. NK, natural killer; TQ, thymoquinone; TC, tumor
cells.

Figure 4.

Production of major cytokines against PC3-RFP cells by NK cells in the presence or absence of TQ. TQ enhances the release of (A) perforin, (B) granzyme B and (C) IFN-α, the major secreted cytokines of NK cells. *P<0.05; **P<0.01; ***P<0.001. NK, natural killer; TQ, thymoquinone; TC, tumor cells.

The correlation between individual cytokines and NK cytotoxicity was analyzed using the Spearman correlation coefficient. Although there was no significant correlation between the cytokine concentrations and NK cytotoxicity; NK cells released higher levels of IFN-α in response to PC3-RFP cells compared to the other cytokines (Fig. 5).

Spearman correlation coefficients
(2-tailed) of perforin, IFN-α and granzyme B cytokines, and natural
killer cytotoxicity with PC3-RFP cells.

Figure 5.

Spearman correlation coefficients (2-tailed) of perforin, IFN-α and granzyme B cytokines, and natural killer cytotoxicity with PC3-RFP cells.

Discussion

Several studies have reported the immunomodulatory effect of TQ on immune cells including NK cells (40–43). The current study focused on investigating the effect of TQ on the cytotoxicity of NK cells and its anticancer activity against PC3-RFP cells. The results demonstrated that treating cancer cells with a high dose of TQ in the presence of NK cells enhanced NK cell cytotoxicity more than NK cells co-cultured with cancer cells alone or in cells with a lower dose of TQ Furthermore, NK cells cultured with cancer cells treated with both TQ concentrations (25 and 50 µM) exhibited considerably increased NK cell cytotoxicity compared with cancer cells treated with TQ alone. This indicates that the antitumor effect of TQ is associated with its stimulation of NK cell function.

Majdalawieh et al (44) assessed how Nigella sativa extract affected the immune systems of mice and reported that the extract activated macrophages, increased NK cell antitumor activity, changed the Th1/Th2 cytokine balance toward a Th1-dominant response, and markedly increased splenocyte proliferation. Despite the fact that TQ, a crucial component of N. sativa, was not isolated, the results indicate that the extract enhances antitumor immunity by inducing both innate and adaptive immunological responses, potentially through TQ (44). Furthermore, a study by Sjs et al (45) produced a nanomedicine based on TQ and reported that administration of the nanomedicine to triple negative breast cancer cell line MDA-MB-231 was associated with apoptosis, DNA damage, slowing down of the cell cycle and prevention of cell division (45). According to these findings, TQ may be a potential strategy for both preventing and treating cancer (46,47).

Nigella sativa may affect cancer pathophysiology via several signaling pathways, such as inducible nitric oxide synthase, p53, ROS, TNF and caspases. These mechanisms have been demonstrated by numerous in vitro and in vivo studies, which suggest that N. sativa concentrates can be used to treat several malignant tumor types at different phases of carcinogenesis (40–42). Additionally, the medicinal plant is well-known for its potent protective benefits against the development and spread of tumors, as well as its anti-inflammatory and immune-stimulating properties, all of which make it a useful supplement to cancer treatment plans. Its impact on the NK cells may be responsible for these effects.

It has been reported that TQ combined with ionizing radiation, such as γ-radiation, has a synergistic lethal effect on breast cancer cells in vitro (42,43,48–50). Moreover, according to the present study, TQ administration directly increased the capacity of NK cells to lyse human PC (PC3-RFP) cells. TQ functions as an antimetabolic medication and may inhibit the development of colorectal cancer carcinogenesis by controlling the glycolytic metabolic pathway and the PI3/AKT axis (51). Additionally, pretreatment with TQ-pH-sensitive liposomes (PSL) decreased cancer marker enzymes, restored the relative weight of the lung and enhanced the activity of antioxidant enzymes in serum, according to a lung cancer study that prepared TQ in a particular formula as TQ-PSL to increase its solubility. Histopathological analysis revealed that TQ-PSL protected lung tissues by decreasing oxidative stress, suppressing inflammatory mediators and inducing apoptosis in pre-cancerous cells (52). TQ itself may be also useful as a treatment for lung adenocarcinoma as it has been reported to inhibit tumor cell proliferation, induce lung cancer cells to undergo apoptosis, markedly reduce TNF and NF-kB activity, and arrest cells in the S phase of the cell cycle (53). TQ has also been widely used in biomaterial treatments (54). Furthermore, TQ has demonstrated notable anti-hepatocellular carcinoma potential, modulating the ERK and p38 signaling pathways, the. Cytotoxic effect of TQ is enhanced by low ERK phosphorylation or ERK inhibition as elevated p-ERK activity protect hepatocellular carcinoma from TQ toxic effect (55,56). TQ and TNF-related apoptosis inducing ligand may cooperate to trigger apoptosis in hepatocellular carcinoma by mediating DNA damage, according to Zhang et al (57) TQ also has an effect on other cancers. For example, TQ markedly increased the amount of reactive oxygen species (ROS) produced by human pancreatic cancer cells whilst simultaneously suppressing the migration and proliferation of cancer cells. Moreover, natural quinones such as TQ may be effective antimetastatic treatments for pancreatic cancer based on these findings (58). As TQ successfully inhibits the fusion of autophagosomes and lysosomes, cancer cells incur apoptosis. Furthermore, TQ causes apoptosis by increasing ROS levels (59). TQ may also function as an immunomodulatory drug that boosts anticancer immune activity, as reported in other in vitro and in vivo research (48,59). It has been also reported that TQ may work in conjunction with chemotherapy and radiation to improve treatment results and reduce treatment side effects (59,60).

Research by Shimasaki et al (61) indicates that NK cells are essential for the regulation of tumor development and metastasis. NK cells contribute to the induction of adaptive anticancer T cell and B cell responses in addition to their function in the early defenses against infection and cancer (61,62). Additionally, NK cells can swiftly eliminate nearby cells that have surface markers associated with neoplastic transformation (53). The direct immune-stimulating impact of TQ on NK is well supported by the current data. In the present study, TQ increased the NK cell production of IFN, which is consistent with other research (4,47) which have reported that the cytotoxic effect of NK cells on tumor cells was increased in the presence of TQ, with increased secretion of perforin, granzyme B, and IFN-α, and that TQ promoted the cytotoxic activity of NK cells against breast cancer MCF-7 cells (63). The results of the present study also supported earlier research by demonstrating the inhibitory action of TQ on human PC PC3-RFP cells. Notably, TQ therapy directly increased the capacity of NK cells to kill PC3-RFP cancer cells. In the NK + TQ + PC3-RFP group in the present study, IFN activity increased, indicating higher concentration. Subsequently, the rise in granzyme B was comparable with earlier research (60,61,64) which showed granzyme B trigger apoptosis by cleaving (BID) BH3 Interacting-Domain Death Agonist pro apoptotic protein of Bcl2 family, causing mitochondrial distribution and activating caspases. This process is crucial for NK cells to induce death in cancer cells (65). Although BID activity was not measured in the present study, previous studies demonstrated the importance of mitochondrial disruption by BID to reach the lethal effect supporting the importance of granzyme B mediated pathways in NK cells cytotoxicity on caspases and causing cell death (64,65). Therefore, granzyme activity measurements are important in studies on NK cytotoxicity.

Furthermore, according to the findings of the present study, TQ affects NK cells, which then directly stimulates the immune system. The TQ-induced immunoreactivity of NK cells against PC3-RFP cells increased when 50 µM TQ was applied, compared with 25 µM. This indicated that the stimulation of NK cells by TQ was associated with its cytotoxic effect on PC3-RFP cells. However, although certain signaling molecules implicated in producing the TQ immunostimulatory action in NK cells have been identified, the specific signaling pathways and molecular targets in these cells are still unknown. It has been suggested that TQ increases NK cell-mediated cytotoxicity by upregulating pro-apoptotic markers such as BAX and BID and downregulating anti-apoptotic proteins such as BCL-2 and myeloid cell leukemia 1 (66). TQ prevents NF-κB, a transcription factor implicated in inflammatory reactions, from activating. TQ also improves NK cell function and lowers pro-inflammatory cytokine expression by inhibiting NF-κB activation, which helps to strengthen antitumor immunity (67). Moreover, by upregulating PTEN, TQ disrupts the PI3K/AKT signaling pathway and reduces AKT activity. This downregulation enhances NK cell-mediated cytotoxicity and promotes apoptosis in cancer cells by reducing cell survival and proliferation signals. Furthermore, antioxidant enzymes are upregulated when TQ stimulates the Nrf2 pathway. By shielding NK cells from oxidative stress, this modulation preserves their functioning and increases their antitumor activity (68). Therefore, more in vitro and in vivo studies are required to identify the target receptors and intracellular and extracellular components that have unique activities in the signal transduction pathways associated with TQ changes in NK cells.

Despite, the promising results of the capacity of TQ to increase NK cell cytotoxicity against PC3-RFP PC cells in vitro, a number of limitations of the present study should be noted. The in vitro design of an experiment cannot fully replicate the intricate interactions in the tumor microenvironment, including tumor-induced immunosuppression, cytokine gradients and immune cell trafficking. In addition, the study did not explore the precise molecular mechanisms behind TQ-induced increased NK cell activity, necessitating further investigation into signaling pathways, receptor expression changes and gene regulatory networks. Moreover, the study used PC3-RFP, a single PC cell line; however, its potential application to other subtypes or primary tumor cells is uncertain due to its heterogeneity. Finally, the observed effects may be influenced by the NK cell source and activation state, with variability in cytotoxic responses potentially introduced by incomplete donor characterization, isolation techniques and baseline NK cell activity. Future therapeutic development depends on determining the ideal concentration range that maximizes tumor cell killing without compromising NK cell viability or function.

In conclusion, the present study demonstrated that TQ, a bioactive substance obtained from Nigella sativa, significantly increases the cytotoxic activity of NK cells against PC3-RFP cells. As a potential supplement to immunotherapeutic approaches in the treatment of PC, the findings imply that TQ may enhance the innate immune response, specifically by increasing NK cell-mediated tumor cell lysis. Furthermore, the potential of TQ as an immunomodulatory agent with anticancer effects is supported by the observed upregulation of NK cell cytotoxicity in its presence. Future research should clarify the precise molecular mechanisms by which TQ improves NK cell function, including its effects on activation receptors, cytokine production and intracellular signaling pathways, in order to build on the present findings. Clinical trials and in vivo research are also necessary to confirm the therapeutic potential of TQ in a physiological setting and evaluate its safety and effectiveness when used in conjunction with currently available immunotherapies. Furthermore, investigating the impact of TQ on different subtypes of PC and its association with the tumor microenvironment may elucidate its suitability for personalized medicine.

Acknowledgements

The authors would like to thank Ms Samar A. Zailaie (King Faisal Specialist Hospital and Research Centre, Jeddah) for technical assistance and scientific advice.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

NIT and NuAA contributed to the study design. NoAA performed the study. NAK and HFA performed the data analysis. NIT, NuAA and HFA performed the data interpretation. HFA and NoAA drafted the manuscript. NIT, NoAA, HFA and NuAA revised the manuscript. All authors critically reviewed the manuscript. NIT and NuAA confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Biomedical Ethics Research Committee of King Abdulaziz University Faculty of Medicine (approval no. No640-20). Written informed consent was obtained from all subjects involved in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Omran AR: The epidemiological transition: A theory of the epidemiology of population change. Millbank Mem Fund Q. 49:509–538. 1971. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Ervik M, Lam F, Laversanne M, Colombet M, Mery L, Piñeros M, Znaor A, Soerjomataram I and Bray F; Global Cancer Observatory, : World. International Agency for Research on Cancer; Lyon, France: https://gco.iarc.who.int/media/globocan/factsheets/populations/900-world-fact-sheet.pdfDecember 26–2024

3 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018.PubMed/NCBI

4 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI

5 

Panigrahi GK, Praharaj PP, Kittaka H, Mridha AR, Black OM, Singh R, Mercer R, van Bokhoven A, Torkko KC, Agarwal C, et al: Exosome proteomic analyses identify inflammatory phenotype and novel biomarkers in African American prostate cancer patients. Cancer Med. 8:1110–1123. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Rawla P: Epidemiology of prostate cancer. World J Oncol. 10:63–89. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Chan JM, Gann PH and Giovannucci EL: Role of diet in prostate cancer development and progression. J Clin Oncol. 23:8152–8160. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Platz EA, Leitzmann MF, Michaud DS, Willett WC and Giovannucci E: Interrelation of energy intake, body size, and physical activity with prostate cancer in a large prospective cohort study. Cancer Res. 63:8542–8548. 2003.PubMed/NCBI

9 

Hegde PS and Chen DS: Top 10 challenges in cancer immunotherapy. Immunity. 52:17–35. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Lopez-Bujanda Z and Drake CG: Myeloid-derived cells in prostate cancer progression: Phenotype and prospective therapies. J Leukoc Biol. 102:393–406. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, Wang Y, Sheikh KL, Terry S, Tagawa ST, et al: Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 1:487–495. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Fridman WH, Pagès F, Sautès-Fridman C and Galon J: The immune contexture in human tumours: Impact on clinical outcome. Nat Rev Cancer. 12:298–306. 2012. View Article : Google Scholar : PubMed/NCBI

13 

O'Donnell JS, Teng MWL and Smyth MJ: Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 16:151–167. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Langer CJ, Gadgeel SM, Borghaei H, Papadimitrakopoulou VA, Patnaik A, Powell SF, Gentzler RD, Martins RG, Stevenson JP, Jalal SI, et al: Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: A randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol. 17:1497–1508. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Patel SH, Rimner A and Cohen RB: Combining immunotherapy and radiation therapy for small cell lung cancer and thymic tumors. Transl Lung Cancer Res. 6:186–195. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Orange JS and Ballas ZK: Natural killer cells in human health and disease. Clin Immunol. 118:1–10. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Vivier E, Tomasello E, Baratin M, Walzer T and Ugolini S: Functions of natural killer cells. Nat Immunol. 9:503–510. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Doherty DG and O'Farrelly C: Innate and adaptive lymphoid cells in the human liver. Immunol Rev. 174:5–20. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Joyce JA and Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 9:239–252. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Ferlazzo G, Pack M, Thomas D, Paludan C, Schmid D, Strowig T, Bougras G, Muller WA, Moretta L and Münz C: Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs. Proc Natl Acad Sci USA. 101:16606–16611. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Romagnani C, Juelke K, Falco M, Morandi B, D'Agostino A, Costa R, Ratto G, Forte G, Carrega P, Lui G, et al: CD56brightCD16- killer Ig-like receptor-NK cells display longer telomeres and acquire features of CD56dim NK cells upon activation. J Immunol. 178:4947–4955. View Article : Google Scholar : PubMed/NCBI

23 

Cooper MA, Fehniger TA and Caligiuri MA: The biology of human natural killer-cell subsets. Trends Immunol. 22:633–640. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Domaica CI, Sierra JM, Zwirner NW and Fuertes MB: Immunomodulation of NK cell activity. Methods Mol Biol. 2097:125–136. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Balsamo M, Scordamaglia F, Pietra G, Manzini C, Cantoni C, Boitano M, Queirolo P, Vermi W, Facchetti F, Moretta A, et al: Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci USA. 106:20847–20852. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Lee HH and Cho H: Improved anti-cancer effect of curcumin on breast cancer cells by increasing the activity of natural killer cells. J Microbiol Biotechnol. 28:874–882. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Montagner IM, Penna A, Fracasso G, Carpanese D, Dalla Pietà A, Barbieri V, Zuccolotto G and Rosato A: Anti-PSMA CAR-engineered NK-92 cells: An off-the-shelf cell therapy for prostate cancer. Cells. 9:13822020. View Article : Google Scholar : PubMed/NCBI

28 

Lundholm M, Schröder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L and Wikström P: Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: Mechanism of immune evasion. PLoS One. 9:e1089252014. View Article : Google Scholar : PubMed/NCBI

29 

Levy EM, Roberti MP and Mordoh J: Natural killer cells in human cancer: From biological functions to clinical applications. Biomed Res Int. 2011:6761982011. View Article : Google Scholar

30 

Tian T and Li Z: Targeting Tim-3 in cancer with resistance to PD-1/PD-L1 blockade. Front Oncol. 11:7311752021. View Article : Google Scholar : PubMed/NCBI

31 

Siemińska I and Baran J: Myeloid-derived suppressor cells as key players and promising therapy targets in prostate cancer. Front Oncol. 12:8624162022. View Article : Google Scholar : PubMed/NCBI

32 

Modena A, Ciccarese C, Iacovelli R, Brunelli M, Montironi R, Fiorentino M, Tortora G and Massari F: Immune checkpoint inhibitors and prostate cancer: A new frontier? Oncol Rev. 10:2932016.PubMed/NCBI

33 

Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, Sato E, Fukuoka S, Tada Y, Tanaka A, et al: PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci USA. 116:9999–10008. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Quatrini L, Mariotti FR, Munari E, Tumino N, Vacca P and Moretta L: The immune checkpoint PD-1 in natural killer cells: Expression, function and targeting in tumour immunotherapy. Cancers (Basel). 12:32852020. View Article : Google Scholar : PubMed/NCBI

35 

Majdalawieh AF and Fayyad MW: Recent advances on the anti-cancer properties of Nigella sativa, a widely used food additive. J Ayurveda Integr Med. 7:173–180. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Dajani EZ, Shahwan TG and Dajani NE: Overview of the preclinical pharmacological properties of Nigella sativa (black seeds): A complementary drug with historical and clinical significance. J Physiol Pharmacol. 67:801–817. 2016.PubMed/NCBI

37 

Bayır AG and Karakaş I: The Role of Nigella sativa and Its Active Component Thymoquinone in Cancer Prevention and Treatment: A Review Article. Eurasian J Med Biol Sci. 1:1–12. 2021.

38 

Ramadan MF: Nutritional value, functional properties and nutraceutical applications of black cumin (Nigella sativa L.): An overview. Int J Food Sci Technol. 42:1208–1218. 2007. View Article : Google Scholar

39 

AlShaibi HF, Ahmed F, Buckle C, Fowles ACM, Awlia J, Cecchini MG and Eaton CL: The BMP antagonist Noggin is produced by osteoblasts in response to the presence of prostate cancer cells. Biotechnol Appl Biochem. 65:407–418. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Randhawa MA and Alghamdi MS: Anticancer activity of Nigella sativa (black seed)-a review. Am J Chin Med. 39:1075–1091. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Schneider-Stock R, Fakhoury IH, Zaki AM, El-Baba CO and Gali-Muhtasib HU: Thymoquinone: Fifty years of success in the battle against cancer models. Drug Discov Today. 19:18–30. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Khan MA, Tania M, Fu S and Fu J: Thymoquinone, as an anticancer molecule: From basic research to clinical investigation. Oncotarget. 8:519072017. View Article : Google Scholar : PubMed/NCBI

43 

Khan MA, Tania M, Wei C, Mei Z, Fu S, Cheng J, Xu J and Fu J: Thymoquinone inhibits cancer metastasis by downregulating TWIST1 expression to reduce epithelial to mesenchymal transition. Oncotarget. 6:19580–19591. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Majdalawieh AF, Hmaidan R and Carr RI: Nigella sativa modulates splenocyte proliferation, Th1/Th2 cytokine profile, macrophage function and NK anti-tumor activity. J Ethnopharmacol. 131:268–275. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Sjs B, Kavithaa K, Poornima A, Haribalan P, Renukadevi B and Sumathi S: Modulation of gene expression by thymoquinone conjugated zinc oxide nanoparticles arrested cell cycle, DNA damage and increased apoptosis in triple negative breast cancer cell line MDA-MB-231. Drug Dev Ind Pharm. 47:1–19. 2022.

46 

Alshaibi HF, Aldarmahi NA, Alkhattabi NA, Alsufiani HM and Tarbiah NI: Studying the anticancer effects of thymoquinone on breast cancer cells through natural killer cell activity. Biomed Res Int. 2022:92186402022. View Article : Google Scholar : PubMed/NCBI

47 

Murphy EM, Centner CS, Bates PJ, Malik MT and Kopechek JA: Delivery of thymoquinone to cancer cells with as1411-conjugated nanodroplets. PLoS One. 15:e02334662020. View Article : Google Scholar : PubMed/NCBI

48 

Peng L, Liu A, Shen Y, Xu HZ, Yang SZ, Ying XZ, Liao W, Liu HX, Lin ZQ, Chen QY, et al: Antitumor and anti-angiogenesis effects of thymoquinone on osteosarcoma through the NF-κB pathway. Oncol Rep. 29:571–578. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Waggoner SN, Daniels KA and Welsh RM: Therapeutic depletion of natural killer cells controls persistent infection. J Virol. 88:1953–1960. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Krebs P, Barnes MJ, Lampe K, Whitley K, Bahjat KS, Beutler B, Janssen E and Hoebe K: NK cell-mediated killing of target cells triggers robust antigen-specific T cell-mediated and humoral responses. Blood. 113:6593–6602. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Karim S, Burzangi AS, Ahmad A, Siddiqui NA, Ibrahim IM, Sharma P, Abualsunun WA and Gabr GA: PI3K-AKT pathway modulation by thymoquinone limits tumor growth and glycolytic metabolism in colorectal cancer. Int J Mol Sci. 23:23052022. View Article : Google Scholar : PubMed/NCBI

52 

Khan A, Alsahli MA, Aljasir MA, Maswadeh H, Mobark MA, Azam F, Allemailem KS, Alrumaihi F, Alhumaydhi FA, Almatroudi AA, et al: Experimental and theoretical insights on chemopreventive effect of the liposomal thymoquinone against benzo [a] pyrene-induced lung cancer in swiss albino mice. J Inflamm Res. 15:2263–2280. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Mirzaei S, Zarrabi A, Hashemi F, Zabolian A, Saleki H, Ranjbar A, Saleh SH, Bagherian M, Sharifzadeh SO, Hushmandi K, et al: Regulation of nuclear factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett. 509:63–80. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Zhang N and Bevan MJ: CD8+ T cells: Foot soldiers of the immune system. Immunity. 35:161–168. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Ibrahim S, Fahim SA, Tadros SA and Badary OA: Suppressive effects of thymoquinone on the initiation stage of diethylnitrosamine hepatocarcinogenesis in rats. J Biochem Mol Toxicol. 36:e230782022. View Article : Google Scholar : PubMed/NCBI

56 

Zhang B, Ting WJ, Gao J, Kang ZF, Huang CY and Weng YJ: Erk phosphorylation reduces the thymoquinone toxicity in human hepatocarcinoma. Environ Toxicol. 36:1990–1998. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Zhang R, Wu T, Zheng P, Liu M, Xu G, Xi M and Yu J: Thymoquinone sensitizes human hepatocarcinoma cells to TRAIL-induced apoptosis via oxidative DNA damage. DNA Repair (Amst). 103:1031172021. View Article : Google Scholar : PubMed/NCBI

58 

Narayanan P, Farghadani R, Nyamathulla S, Rajarajeswaran J, Thirugnanasampandan R and Bhuwaneswari G: Natural quinones induce ROS-mediated apoptosis and inhibit cell migration in PANC-1 human pancreatic cancer cell line. J Biochem Mol Toxicol. 36:e230082022. View Article : Google Scholar : PubMed/NCBI

59 

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. 320:1090222020. View Article : Google Scholar : PubMed/NCBI

60 

Sutton VR, Davis JE, Cancilla M, Johnstone RW, Ruefli AA, Sedelies K, Browne KA and Trapani JA: Initiation of apoptosis by granzyme B requires direct cleavage of bid, but not direct granzyme B-mediated caspase activation. J Exp Med. 192:1403–1414. 2000. View Article : Google Scholar : PubMed/NCBI

61 

Shimasaki N, Jain A and Campana D: NK cells for cancer immunotherapy. Nat Rev Drug Discov. 19:200–218. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, Takeda K, van Dommelen SL, Degli-Esposti MA and Hayakawa Y: Activation of NK cell cytotoxicity. Mol Immunol. 42:501–510. 2005. View Article : Google Scholar : PubMed/NCBI

63 

Kingdom of Saudi Arabia, Saudi Health Council, National Cancer Center, Saudi Cancer Registry, . Cancer Incididence Report Saudi Arabia 2020. https://shc.gov.sa/Arabic/NewNCC/Activities/AnnualReports/2020.pdfJanuary 5–2025

64 

Chowdhury D and Lieberman J: Death by a thousand cuts: Granzyme pathways of programmed cell death. Annu Rev Immunol. 26:389–420. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Young JD, Hengartner H, Podack ER and Cohn ZA: Purification and characterization of a cytolytic pore-forming protein from granules of cloned lymphocytes with natural killer activity. Cell. 44:849–859. 1986. View Article : Google Scholar : PubMed/NCBI

66 

Singh SK, Mishra MK, Lillard JW and Singh R: Thymoquinone enhanced the tumoricidal activity of NK cells against lung cancer. J Immunol. 200 (Supplement_1):S124–S125. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Sethi G, Ahn KS and Aggarwal BB: Targeting nuclear factor-κB activation pathway by thymoquinone: role in suppression of antiapoptotic gene products and enhancement of apoptosis. Mol Cancer Res. 6:1059–1070. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Sadeghi E, Imenshahidi M and Hosseinzadeh H: Molecular mechanisms and signaling pathways of black cumin (Nigella sativa) and its active constituent, thymoquinone: A review. Mol Biol Rep. 50:5439–5454. 2023. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Aldarmahi NA, Tarbiah NI, Alkhattabi NA and Alshaibi HF: Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity. Oncol Lett 30: 530, 2025.
APA
Aldarmahi, N.A., Tarbiah, N.I., Alkhattabi, N.A., & Alshaibi, H.F. (2025). Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity. Oncology Letters, 30, 530. https://doi.org/10.3892/ol.2025.15276
MLA
Aldarmahi, N. A., Tarbiah, N. I., Alkhattabi, N. A., Alshaibi, H. F."Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity". Oncology Letters 30.5 (2025): 530.
Chicago
Aldarmahi, N. A., Tarbiah, N. I., Alkhattabi, N. A., Alshaibi, H. F."Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity". Oncology Letters 30, no. 5 (2025): 530. https://doi.org/10.3892/ol.2025.15276
Copy and paste a formatted citation
x
Spandidos Publications style
Aldarmahi NA, Tarbiah NI, Alkhattabi NA and Alshaibi HF: Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity. Oncol Lett 30: 530, 2025.
APA
Aldarmahi, N.A., Tarbiah, N.I., Alkhattabi, N.A., & Alshaibi, H.F. (2025). Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity. Oncology Letters, 30, 530. https://doi.org/10.3892/ol.2025.15276
MLA
Aldarmahi, N. A., Tarbiah, N. I., Alkhattabi, N. A., Alshaibi, H. F."Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity". Oncology Letters 30.5 (2025): 530.
Chicago
Aldarmahi, N. A., Tarbiah, N. I., Alkhattabi, N. A., Alshaibi, H. F."Anticancer effects of thymoquinone on prostate cancer cells and natural killer cell activity". Oncology Letters 30, no. 5 (2025): 530. https://doi.org/10.3892/ol.2025.15276
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team