1
|
Majdalawieh AF, Al-Samaraie S, Terro TM. Molecular Mechanisms and Signaling Pathways Underlying the Therapeutic Potential of Thymoquinone Against Colorectal Cancer. Molecules 2024; 29:5907. [PMID: 39769996 PMCID: PMC11679644 DOI: 10.3390/molecules29245907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Thymoquinone (TQ), a bioactive compound derived from Nigella sativa, has garnered significant attention for its potential as a natural anti-cancer agent, particularly in the context of colorectal cancer. This review provides a detailed synthesis of the current literature on the anti-cancer properties of TQ in colorectal cancer cells, exploring both in vitro and in vivo studies to elucidate its mechanisms of action. TQ effectively induces apoptosis, inhibits cell proliferation, and reduces metastasis in colorectal cancer cells by modulating key molecular pathways such as PI3K/AKT/mTOR, NF-κB, STAT3, and MAPK. It causes mitochondrial dysfunction and activates caspases, contributing to its pro-apoptotic effects. TQ also regulates EMT and targets cancer stem cells, reducing the likelihood of metastasis. Moreover, its antioxidant properties contribute to its protective role against cancer progression. While preclinical studies provide strong evidence of TQ's efficacy, further clinical studies are essential to establish its therapeutic potential in humans. This review underscores TQ's promising role as a natural agent with the potential to significantly improve colorectal cancer treatment outcomes.
Collapse
Affiliation(s)
- Amin F. Majdalawieh
- Department of Biology, Chemistry, and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Saud Al-Samaraie
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
| | - Tala M. Terro
- Department of Biology, Chemistry, and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| |
Collapse
|
2
|
Deng X, Yang Z, Chan KW, Abu Bakar MZ. Exploring the Therapeutic Potential of 5-Fluorouracil-Loaded Calcium Carbonate Nanoparticles Combined with Natural Compound Thymoquinone for Colon Cancer Treatment. Pharmaceutics 2024; 16:1011. [PMID: 39204357 PMCID: PMC11360259 DOI: 10.3390/pharmaceutics16081011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Given the need for novel and effective therapies for colon cancer, this study aimed to investigate the effects of 5-fluorouracil-loaded calcium carbonate nanoparticles (5FU-CaCO3np) combined with thymoquinone (TQ) against colon cancer. A shaking incubator and a high-speed homogenizer were used to prepare the optimal 5FU-CaCO3np, with characterizations of physicochemical properties, in vitro drug release profile, and biocompatibility. In vitro experiments and molecular docking were employed to evaluate the therapeutic potential of the combination for colon cancer treatment. Study results revealed that 5FU-CaCO3np with a size of approximately 130 nm was synthesized using the high-speed homogenizer. Its favorable biocompatibility, pH sensitivity, and sustained release properties facilitated reduced toxic side effects of 5-FU on NIH3T3 normal cells and enhanced inhibitory effects on CT26 colon cancer cells. The combination of 5FU-CaCO3np (1.875 μM) and TQ (30 μM) showed significantly superior anti-colon cancer effects to 5FU-CaCO3np alone in terms of cell proliferation and migration inhibition, cell apoptosis induction, and spheroid growth suppression in CT26 cells (p < 0.05), with strong interactions between the drugs and targets (E-cadherin, Bcl-2, PCNA, and MMP-2). These results provide evidence for 5FU-CaCO3np as a novel regimen against colon cancer. Combining 5FU-CaCO3np and TQ may offer a new perspective for colon cancer therapy.
Collapse
Affiliation(s)
- Xi Deng
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (X.D.); (Z.Y.); (K.W.C.)
| | - Zhongming Yang
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (X.D.); (Z.Y.); (K.W.C.)
| | - Kim Wei Chan
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (X.D.); (Z.Y.); (K.W.C.)
| | - Md Zuki Abu Bakar
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (X.D.); (Z.Y.); (K.W.C.)
- Department of Veterinary Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
3
|
Abutayeh RF, Altah M, Mehdawi A, Al-Ataby I, Ardakani A. Chemopreventive Agents from Nature: A Review of Apigenin, Rosmarinic Acid, and Thymoquinone. Curr Issues Mol Biol 2024; 46:6600-6619. [PMID: 39057035 PMCID: PMC11276303 DOI: 10.3390/cimb46070393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer, a major challenge to global health and healthcare systems, requires the study of alternative and supportive treatments due to the limitations of conventional therapies. This review examines the chemopreventive potential of three natural compounds: rosmarinic acid, apigenin, and thymoquinone. Derived from various plants, these compounds have demonstrated promising chemopreventive properties in in vitro, in vivo, and in silico studies. Specifically, they have been shown to inhibit cancer cell growth, induce apoptosis, and modulate key signaling pathways involved in cancer progression. The aim of this review is to provide a comprehensive overview of the current research on these phytochemicals, elucidating their mechanisms of action, therapeutic efficacy, and potential as adjuncts to traditional cancer therapies. This information serves as a valuable resource for researchers and healthcare providers interested in expanding their knowledge within the field of alternative cancer therapies.
Collapse
Affiliation(s)
- Reem Fawaz Abutayeh
- Faculty of Pharmacy, Applied Science Private University, Amman 11937, Jordan;
| | - Maram Altah
- School of Pharmacy, Al-Qadisiyah College, Amman 11118, Jordan;
| | - Amani Mehdawi
- School of Pharmacy, Al-Qadisiyah College, Amman 11118, Jordan;
| | - Israa Al-Ataby
- Faculty of Pharmacy, Applied Science Private University, Amman 11937, Jordan;
| | - Adel Ardakani
- College of Pharmacy, Amman Arab University, Amman 11953, Jordan;
| |
Collapse
|
4
|
Jassi C, Kuo WW, Chang YC, Wang TF, Li CC, Ho TJ, Hsieh DJY, Kuo CH, Chen MC, Huang CY. Aloin and CPT-11 combination activates miRNA-133b and downregulates IGF1R- PI3K/AKT/mTOR and MEK/ERK pathways to inhibit colorectal cancer progression. Biomed Pharmacother 2023; 169:115911. [PMID: 38000359 DOI: 10.1016/j.biopha.2023.115911] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023] Open
Abstract
CPT-11 is one of the drugs employed in colorectal cancer treatment and has faced challenges in the form of resistance. The insulin-like growth factor 1 receptor is a tyrosine kinase receptor that mediates cancer cell survival and drug resistance. It is frequently overexpressed in colorectal cancer and has previously been identified as a microRNA target. MicroRNAs are non-coding RNA molecules that regulate gene function by suppressing messenger RNA translation. Studies have demonstrated that natural compounds can regulate microRNA function and their target genes. Therefore, combining natural compounds with existing cancer drugs can enhance the therapeutic efficacy. We investigated a natural compound, Aloin, for the potential sensitization of colorectal cancer to CPT-11. We used western blot, MTT cell viability assay, flow cytometry, and microRNA/gene knockdown and overexpression experiments, as well as an in vivo mouse model. Our investigation revealed that combining Aloin with CPT-11 exerts an enhanced anti-tumor effect in colorectal cancer. This combination reduced cell viability and induced apoptosis, both in vivo and in vitro. Furthermore, this combination upregulated miRNA-133b, while downregulating the IGF1R and its downstream MEK/ERK, and PI3K/AKT/mTOR pathways. Our findings suggests that CPT-11 and Aloin are potential combination treatment partners against colorectal cancer. MicroRNA-133b may serve as a co-therapeutic target with IGF1R against colorectal cancer, which might overcome the existing treatment limitations.
Collapse
Affiliation(s)
- Chikondi Jassi
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yu-Chun Chang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan; School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
| | - Chi-Cheng Li
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan; Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jung Ho
- Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan; Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Ming-Cheng Chen
- Department of Surgery, Division of Colorectal Surgery, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria related diseases research center, Hualien Tzu Chi Hospital, Hualien 970, Taiwan; Graduate Institute of Biomedicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
5
|
Kurowska N, Madej M, Strzalka-Mrozik B. Thymoquinone: A Promising Therapeutic Agent for the Treatment of Colorectal Cancer. Curr Issues Mol Biol 2023; 46:121-139. [PMID: 38248312 PMCID: PMC10814900 DOI: 10.3390/cimb46010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and is responsible for approximately one million deaths each year. The current standard of care is surgical resection of the lesion and chemotherapy with 5-fluorouracil (5-FU). However, of concern is the increasing incidence in an increasingly younger patient population and the ability of CRC cells to develop resistance to 5-FU. In this review, we discuss the effects of thymoquinone (TQ), one of the main bioactive components of Nigella sativa seeds, on CRC, with a particular focus on the use of TQ in combination therapy with other chemotherapeutic agents. TQ exhibits anti-CRC activity by inducing a proapoptotic effect and inhibiting proliferation, primarily through its effect on the regulation of signaling pathways crucial for tumor progression and oxidative stress. TQ can be used synergistically with chemotherapeutic agents to enhance their anticancer effects and to influence the expression of signaling pathways and other genes important in cancer development. These data appear to be most relevant for co-treatment with 5-FU. We believe that TQ is a suitable candidate for consideration in the chemoprevention and adjuvant therapy for CRC, but further studies, including clinical trials, are needed to confirm its safety and efficacy in the treatment of cancer.
Collapse
Affiliation(s)
- Natalia Kurowska
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (N.K.); (M.M.)
| | - Marcel Madej
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (N.K.); (M.M.)
- Silesia LabMed, Centre for Research and Implementation, Medical University of Silesia, 40-752 Katowice, Poland
| | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (N.K.); (M.M.)
| |
Collapse
|
6
|
Sheikhnia F, Rashidi V, Maghsoudi H, Majidinia M. Potential anticancer properties and mechanisms of thymoquinone in colorectal cancer. Cancer Cell Int 2023; 23:320. [PMID: 38087345 PMCID: PMC10717210 DOI: 10.1186/s12935-023-03174-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/04/2023] [Indexed: 10/14/2024] Open
Abstract
Colorectal neoplasms are one of the deadliest diseases among all cancers worldwide. Thymoquinone (TQ) is a natural compound of Nigella sativa that has been used in traditional medicine against a variety of acute/chronic diseases such as asthma, bronchitis, rheumatism, headache, back pain, anorexia, amenorrhea, paralysis, inflammation, mental disability, eczema, obesity, infections, depression, dysentery, hypertension, gastrointestinal, cardiovascular, hepatic, and renal disorders. This review aims to present a detailed report on the studies conducted on the anti-cancer properties of TQ against colorectal cancer, both in vitro and in vivo. TQ stands as a promising natural therapeutic agent that can enhance the efficacy of existing cancer treatments while minimizing the associated adverse effects. The combination of TQ with other anti-neoplastic agents promoted the efficacy of existing cancer treatments. Further research is needed to acquire a more comprehensive understanding of its exact molecular targets and pathways and maximize its clinical usefulness. These investigations may potentially aid in the development of novel techniques to combat drug resistance and surmount the obstacles presented by chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
7
|
Yan CF, Xia J, Qun WS, Bing WY, Guo WJ, Yong HG, Sheng SJ, Lei ZG. Tumor-associated macrophages-derived exo-let-7a promotes osteosarcoma metastasis via targeting C15orf41 in osteosarcoma. ENVIRONMENTAL TOXICOLOGY 2023; 38:1318-1331. [PMID: 36919336 DOI: 10.1002/tox.23766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Osteosarcoma (OS) immune environment is complexed and the immune factors-related to OS progression need to be explored. Tumor-associated macrophages (TAMs) are regarded as immune suppressive and tumor-promoting cells. However, the underlying mechanisms through which TAMs function are still fragmentary. Here, we aim to explore the underlying mechanisms by which TAMs regulate OS progression. METHODS TAMs from OS tissues were isolated by flow cytometry. Exosomes derived from TAMs were separated using ultracentrifugation and western blotting. Transmission electron microscopy (TEM), and flow cytometry were constructed to characterize TAMs-derived exosomes. Additionally, the differential MicroRNAs (miRNAs) and genes were detected through RNA sequencing, and further validated using real-time PCR (RT-PCR). OS cell metastasis ability was assessed using transwell invasion and scratch wound healing assays. MiRNAs mimic and lentiviral vectors were utilized to explore the effects on OS progression. RESULTS Exosome secreted by TAMs accelerated the OS metastasis. Let-7a level was upregulated in TAMs derived exosomes, which downregulated C15orf41 by targeting 3'-untranslated region (UTR). Furthermore, overexpressing let-7a enhanced invasion and migration by blocking the transcription of C15orf41. In consistent, up-regulating let-7a promoted OS progression and made the prognosis to be worse, which can be reversed by C15orf41 overexpression. CONCLUSION This study highlighted the critical role of TAMs-derived exosomes in OS progression and explored the potential value of the let-7a/C15orf41 axis as an indicator or target for OS.
Collapse
Affiliation(s)
- Chen-Fei Yan
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wang-Si Qun
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei-Yi Bing
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wu-Jian Guo
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Huang-Gang Yong
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Jing Sheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhao-Guang Lei
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Eid EEM, Almaiman AA, Alshehade SA, Alsalemi W, Kamran S, Suliman FO, Alshawsh MA. Characterization of Thymoquinone-Sulfobutylether-β-Cyclodextrin Inclusion Complex for Anticancer Applications. Molecules 2023; 28:4096. [PMID: 37241838 PMCID: PMC10223034 DOI: 10.3390/molecules28104096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Thymoquinone (TQ) is a quinone derived from the black seed Nigella sativa and has been extensively studied in pharmaceutical and nutraceutical research due to its therapeutic potential and pharmacological properties. Although the chemopreventive and potential anticancer effects of TQ have been reported, its limited solubility and poor delivery remain the major limitations. In this study, we aimed to characterize the inclusion complexes of TQ with Sulfobutylether-β-cyclodextrin (SBE-β-CD) at four different temperatures (293-318 K). Additionally, we compared the antiproliferative activity of TQ alone to TQ complexed with SBE-β-CD on six different cancer cell lines, including colon, breast, and liver cancer cells (HCT-116, HT-29, MDA-MB-231, MCF-7, SK-BR-3, and HepG2), using an MTT assay. We calculated the thermodynamic parameters (ΔH, ΔS, and ΔG) using the van't Holf equation. The inclusion complexes were characterized by X-ray diffraction (XRD), Fourier transforms infrared (FT-IR), and molecular dynamics using the PM6 model. Our findings revealed that the solubility of TQ was improved by ≥60 folds, allowing TQ to penetrate completely into the cavity of SBE-β-CD. The IC50 values of TQ/SBE-β-CD ranged from 0.1 ± 0.01 µg/mL against SK-BR-3 human breast cancer cells to 1.2 ± 0.16 µg/mL against HCT-116 human colorectal cancer cells, depending on the cell line. In comparison, the IC50 values of TQ alone ranged from 0.2 ± 0.01 µg/mL to 4.7 ± 0.21 µg/mL. Overall, our results suggest that SBE-β-CD can enhance the anticancer effect of TQ by increasing its solubility and bioavailability and cellular uptake. However, further studies are necessary to fully understand the underlying mechanisms and potential side effects of using SBE-β-CD as a drug delivery system for TQ.
Collapse
Affiliation(s)
- Eltayeb E. M. Eid
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Unaizah 51911, Saudi Arabia
| | - Amer A. Almaiman
- Unit of Scientific Research, Applied College, Qassim University, Unaizah 51911, Saudi Arabia
| | | | - Wardah Alsalemi
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Sareh Kamran
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - FakhrEldin O. Suliman
- Department of Chemistry, College of Science, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| | | |
Collapse
|
9
|
Inhibition of RNA Polymerase III Augments the Anti-Cancer Properties of TNFα. Cancers (Basel) 2023; 15:cancers15051495. [PMID: 36900285 PMCID: PMC10000776 DOI: 10.3390/cancers15051495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Tumour necrosis factor alpha (TNFα) is a multifunctional cytokine that plays a pivotal role in apoptosis, cell survival, as well as in inflammation and immunity. Although named for its antitumor properties, TNFα also has tumour-promoting properties. TNFα is often present in large quantities in tumours, and cancer cells frequently acquire resistance to this cytokine. Consequently, TNFα may increase the proliferation and metastatic potential of cancer cells. Furthermore, the TNFα-driven increase in metastasis is a result of the ability of this cytokine to induce the epithelial-to-mesenchymal transition (EMT). Overcoming the resistance of cancer cells to TNFα may have a potential therapeutic benefit. NF-κB is a crucial transcription factor mediating inflammatory signals and has a wide-ranging role in tumour progression. NF-κB is strongly activated in response to TNFα and contributes to cell survival and proliferation. The pro-inflammatory and pro-survival function of NF-κB can be disrupted by blocking macromolecule synthesis (transcription, translation). Consistently, inhibition of transcription or translation strongly sensitises cells to TNFα-induced cell death. RNA polymerase III (Pol III) synthesises several essential components of the protein biosynthetic machinery, such as tRNA, 5S rRNA, and 7SL RNA. No studies, however, directly explored the possibility that specific inhibition of Pol III activity sensitises cancer cells to TNFα. Here we show that in colorectal cancer cells, Pol III inhibition augments the cytotoxic and cytostatic effects of TNFα. Pol III inhibition enhances TNFα-induced apoptosis and also blocks TNFα-induced EMT. Concomitantly, we observe alterations in the levels of proteins related to proliferation, migration, and EMT. Finally, our data show that Pol III inhibition is associated with lower NF-κB activation upon TNFα treatment, thus potentially suggesting the mechanism of Pol III inhibition-driven sensitisation of cancer cells to this cytokine.
Collapse
|
10
|
Plant-Derived Bioactive Compounds in Colorectal Cancer: Insights from Combined Regimens with Conventional Chemotherapy to Overcome Drug-Resistance. Biomedicines 2022; 10:biomedicines10081948. [PMID: 36009495 PMCID: PMC9406120 DOI: 10.3390/biomedicines10081948] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Acquired drug resistance represents a major clinical problem and one of the biggest limitations of chemotherapeutic regimens in colorectal cancer. Combination regimens using standard chemotherapeutic agents, together with bioactive natural compounds derived from diet or plants, may be one of the most valuable strategies to overcome drug resistance and re-sensitize chemoresistant cells. In this review, we highlight the effect of combined regimens based on conventional chemotherapeutics in conjunction with well-tolerated plant-derived bioactive compounds, mainly curcumin, resveratrol, and EGCG, with emphasis on the molecular mechanisms associated with the acquired drug resistance.
Collapse
|
11
|
Hassanzadeh P, Arbabi E. Presenting a bioactive nanotherapeutic agent for colon cancer treatment. Eur J Pharmacol 2022; 927:175084. [PMID: 35679890 DOI: 10.1016/j.ejphar.2022.175084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/03/2022]
Abstract
Colon cancer (CC) is one of the major causes of death worldwide. Insufficient drug concentration, non-specificity, or serious adverse effects of the conventional chemotherapeutic agents necessitate application of more effective treatment options. Herein, poly-ursolic acid, a polymer with anticancer effect, has been self-assembled for producing nanoparticles (NPs) for delivery of irinotecan (IRN) which is usually associated with poor solubility and severe adverse effects. NPs showed therapeutic efficiency by themselves in vivo and in vitro. IRN-loaded NPs with appropriate physicochemical characteristics, released IRN in a controlled fashion and demonstrated more efficient cytotoxicity, lower clearance rate and distribution volume, higher Cmax and AUC, prolonged t1/2, increased accumulation in tumor, and therapeutic effects in vivo as compared to free drug. There was no significant alteration of body weight or damage to the major organs. The prepared bioactive nanoplatform via improvement of IRN efficiency could be applied for inducing synergistic toxicity against CC.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 13169-43551, Iran; Sasan Hospital, Tehran, 14159-83391, Iran.
| | - Elham Arbabi
- Research Center for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone. Int J Mol Sci 2022; 23:ijms23116311. [PMID: 35682990 PMCID: PMC9181073 DOI: 10.3390/ijms23116311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
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.
Collapse
|
13
|
Tabassum S, Thakur V, Rosli N, Ichwan SJA, Mishra P, Suriyah WH. Therapeutic implications of thymoquinone and its molecular and functional mechanisms against oral and lung cancer. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14
|
Al Bitar S, Ballout F, Monzer A, Kanso M, Saheb N, Mukherji D, Faraj W, Tawil A, Doughan S, Hussein M, Abou-Kheir W, Gali-Muhtasib H. Thymoquinone Radiosensitizes Human Colorectal Cancer Cells in 2D and 3D Culture Models. Cancers (Basel) 2022; 14:1363. [PMID: 35326517 PMCID: PMC8945905 DOI: 10.3390/cancers14061363] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Resistance of cancer cells and normal tissue toxicity of ionizing radiation (IR) are known to limit the success of radiotherapy. There is growing interest in using IR with natural compounds to sensitize cancer cells and spare healthy tissues. Thymoquinone (TQ) was shown to radiosensitize several cancers, yet no studies have investigated its radiosensitizing effects on colorectal cancer (CRC). Here, we combined TQ with IR and determined its effects in two-dimensional (2D) and three-dimensional (3D) culture models derived from HCT116 and HT29 CRC cells, and in patient-derived organoids (PDOs). TQ sensitized CRC cells to IR and reduced cell viability and clonogenic survival and was non-toxic to non-tumorigenic intestinal cells. TQ sensitizing effects were associated with G2/M arrest and DNA damage as well as changes in key signaling molecules involved in this process. Combining a low dose of TQ (3 µM) with IR (2 Gy) inhibited sphere formation by 100% at generation 5 and this was associated with inhibition of stemness and DNA repair. These doses also led to ~1.4- to ~3.4-fold decrease in organoid forming ability of PDOs. Our findings show that combining TQ and IR could be a promising therapeutic strategy for eradicating CRC cells.
Collapse
Affiliation(s)
- Samar Al Bitar
- Department of Biology, American University of Beirut, Beirut 1107-2020, Lebanon; (S.A.B.); (F.B.); (A.M.)
| | - Farah Ballout
- Department of Biology, American University of Beirut, Beirut 1107-2020, Lebanon; (S.A.B.); (F.B.); (A.M.)
| | - Alissar Monzer
- Department of Biology, American University of Beirut, Beirut 1107-2020, Lebanon; (S.A.B.); (F.B.); (A.M.)
| | - Mariam Kanso
- Division of General Surgery, Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (M.K.); (W.F.); (S.D.); (M.H.)
| | - Nour Saheb
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (N.S.); (A.T.)
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon;
| | - Walid Faraj
- Division of General Surgery, Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (M.K.); (W.F.); (S.D.); (M.H.)
| | - Ayman Tawil
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (N.S.); (A.T.)
| | - Samer Doughan
- Division of General Surgery, Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (M.K.); (W.F.); (S.D.); (M.H.)
| | - Maher Hussein
- Division of General Surgery, Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon; (M.K.); (W.F.); (S.D.); (M.H.)
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hala Gali-Muhtasib
- Department of Biology, American University of Beirut, Beirut 1107-2020, Lebanon; (S.A.B.); (F.B.); (A.M.)
| |
Collapse
|
15
|
Homayoonfal M, Asemi Z, Yousefi B. Potential anticancer properties and mechanisms of thymoquinone in osteosarcoma and bone metastasis. Cell Mol Biol Lett 2022; 27:21. [PMID: 35236304 PMCID: PMC8903697 DOI: 10.1186/s11658-022-00320-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
Despite great advances, therapeutic approaches of osteosarcoma, the most prevalent class of preliminary pediatric bone tumors, as well as bone-related malignancies, continue to demonstrate insufficient adequacy. In recent years, a growing trend toward applying natural bioactive compounds, particularly phytochemicals, as novel agents for cancer treatment has been observed. Bioactive phytochemicals exert their anticancer features through two main ways: they induce cytotoxic effects against cancerous cells without having any detrimental impact on normal cell macromolecules such as DNA and enzymes, while at the same time combating the oncogenic signaling axis activated in tumor cells. Thymoquinone (TQ), the most abundant bioactive compound of Nigella sativa, has received considerable attention in cancer treatment owing to its distinctive properties, including apoptosis induction, cell cycle arrest, angiogenesis and metastasis inhibition, and reactive oxygen species (ROS) generation, along with inducing immune system responses and reducing side effects of traditional chemotherapeutic drugs. The present review is focused on the characteristics and mechanisms by which TQ exerts its cytotoxic effects on bone malignancies.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
de Klerk DJ, de Keijzer MJ, Dias LM, Heemskerk J, de Haan LR, Kleijn TG, Franchi LP, Heger M. Strategies for Improving Photodynamic Therapy Through Pharmacological Modulation of the Immediate Early Stress Response. Methods Mol Biol 2022; 2451:405-480. [PMID: 35505025 DOI: 10.1007/978-1-0716-2099-1_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.
Collapse
Affiliation(s)
- Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Faculdade de Ciências da Saúde (FCS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Jordi Heemskerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Sciences, and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Zhang B, Ting W, Gao J, Kang Z, Huang C, Weng Y. Erk phosphorylation reduces the thymoquinone toxicity in human hepatocarcinoma. ENVIRONMENTAL TOXICOLOGY 2021; 36:1990-1998. [PMID: 34173702 PMCID: PMC8456969 DOI: 10.1002/tox.23317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/11/2021] [Accepted: 06/15/2021] [Indexed: 05/05/2023]
Abstract
Although enormous achievements have been made in targeted molecular therapies against hepatocellular carcinoma (HCC), the treatments can only prolong the life of patients with extrahepatic metastases. We evaluated thymoquinone (TQ), a compound from Nigella sativa Linn., for its anti-cancer effect on SK-Hep1 cells and HCC-xenograft nude mice. TQ effectively triggered cell death and activated p38 and extracellular signal-regulated kinases (Erk) pathways up to 24 h after treatment in cells. TQ-induced cell death was reversed by p38 inhibitor; however, it was enhanced by si-Erk. The caspase3 activation and TUNEL assay revealed a stronger toxic effect upon co-treatment with TQ and si-Erk. Our study suggested that phosphorylation of p38 in SK-Hep1 cells constituted the major factor leading to cell apoptosis, whereas phosphorylation of Erk led to drug resistance. Furthermore, TQ therapeutic effect was improved upon Erk inhibition in HCC-xenograft nude mice. TQ could present excellent anti-HCC potential under suitable p-Erk inhibiting conditions.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Hepatobiliary SurgeryThe Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuanChina
| | - Wei‐Jen Ting
- Basic Medical Science LaboratoryThe Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuanChina
| | - Jun Gao
- Basic Medical Science LaboratoryThe Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuanChina
| | - Zhan‐Fang Kang
- Basic Medical Science LaboratoryThe Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuanChina
| | - Chih‐Yang Huang
- Graduate Institute of Biomedical ScienceChina Medical UniversityTaichungTaiwan
- Cardiovascular and Mitochondrial Related Disease Research CenterHualien Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationHualienTaiwan
- Department of Medical ResearchChina Medical University Hospital, China Medical UniversityTaichungTaiwan
- Center of General EducationBuddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and TechnologyHualienTaiwan
| | - Yi‐Jiun Weng
- Basic Medical Science LaboratoryThe Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuanChina
| |
Collapse
|
18
|
Apoptotic and Non-Apoptotic Modalities of Thymoquinone-Induced Lymphoma Cell Death: Highlight of the Role of Cytosolic Calcium and Necroptosis. Cancers (Basel) 2021; 13:cancers13143579. [PMID: 34298792 PMCID: PMC8304872 DOI: 10.3390/cancers13143579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 01/17/2023] Open
Abstract
Simple Summary Diffuse large B cell lymphoma (DLBCL) represents the most common type of non-Hodgkin lymphoma with a high curability rate. However, 40% of patients will relapse or exhibit refractory disease, and compromised apoptotic pathways is among the prognosis-worsening factors. Therefore, drugging non-apoptotic modalities might be therapeutically promising. Thymoquinone (TQ) has been reported to promote apoptosis in cancer cells. Herein, we show that TQ selectively kills DLBCL cells, either cell lines or primary lymphoma cells bearing resistance features to standard treatment. Investigations show that, although TQ induced apoptotic markers, non-apoptotic death was the major mechanism responsible for TQ-induced cellular demise. We demonstrate critical and selective roles of cytosolic calcium and necroptosis in TQ-induced non-apoptotic cell death. Finally, TQ exhibits an improved selectivity profile over conventional chemotherapy. Collectively, this work provides new insights into the mode of action of TQ and points to the therapeutic relevance of non-apoptotic modalities as a fail-safe mechanism for pro-apoptotic DLBCL therapies. Abstract Targeting non-apoptotic modalities might be therapeutically promising in diffuse large B cell lymphoma (DLBCL) patients with compromised apoptotic pathways. Thymoquinone (TQ) has been reported to promote apoptosis in cancer cells, but little is known about its effect on non-apoptotic pathways. This work investigates TQ selectivity against DLBCL cell lines and the cell death mechanisms. TQ reduces cell viability and kills cell lines with minimal toxicity on normal hematological cells. Mechanistically, TQ promotes the mitochondrial caspase pathway and increases genotoxicity. However, insensitivity of most cell lines to caspase inhibition by z-VAD-fmk (benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone) pointed to a critical role of non-apoptotic signaling. In cells dying through non-apoptotic death, TQ increases endoplasmic reticulum (ER) stress markers and substantially increases cytosolic calcium ([Ca2+]c) through ER calcium depletion and activation of store-operated calcium entry (SOCE). Chelation of [Ca2+]c, but not SOCE inhibitors, reduces TQ-induced non-apoptotic cell death, highlighting the critical role of calcium in a non-apoptotic effect of TQ. Investigations showed that TQ-induced [Ca2+]c signaling is primarily initiated by necroptosis upstream to SOCE, and inhibition necroptosis by necrostatin-1 alone or with z-VAD-fmk blocks the cell death. Finally, TQ exhibits an improved selectivity profile over standard chemotherapy agents, suggesting a therapeutic relevance of the pro-necroptotic effect of TQ as a fail-safe mechanism for DLBCL therapies targeting apoptosis.
Collapse
|
19
|
Hannan MA, Rahman MA, Sohag AAM, Uddin MJ, Dash R, Sikder MH, Rahman MS, Timalsina B, Munni YA, Sarker PP, Alam M, Mohibbullah M, Haque MN, Jahan I, Hossain MT, Afrin T, Rahman MM, Tahjib-Ul-Arif M, Mitra S, Oktaviani DF, Khan MK, Choi HJ, Moon IS, Kim B. Black Cumin ( Nigella sativa L.): A Comprehensive Review on Phytochemistry, Health Benefits, Molecular Pharmacology, and Safety. Nutrients 2021; 13:1784. [PMID: 34073784 PMCID: PMC8225153 DOI: 10.3390/nu13061784] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Mounting evidence support the potential benefits of functional foods or nutraceuticals for human health and diseases. Black cumin (Nigella sativa L.), a highly valued nutraceutical herb with a wide array of health benefits, has attracted growing interest from health-conscious individuals, the scientific community, and pharmaceutical industries. The pleiotropic pharmacological effects of black cumin, and its main bioactive component thymoquinone (TQ), have been manifested by their ability to attenuate oxidative stress and inflammation, and to promote immunity, cell survival, and energy metabolism, which underlie diverse health benefits, including protection against metabolic, cardiovascular, digestive, hepatic, renal, respiratory, reproductive, and neurological disorders, cancer, and so on. Furthermore, black cumin acts as an antidote, mitigating various toxicities and drug-induced side effects. Despite significant advances in pharmacological benefits, this miracle herb and its active components are still far from their clinical application. This review begins with highlighting the research trends in black cumin and revisiting phytochemical profiles. Subsequently, pharmacological attributes and health benefits of black cumin and TQ are critically reviewed. We overview molecular pharmacology to gain insight into the underlying mechanism of health benefits. Issues related to pharmacokinetic herb-drug interactions, drug delivery, and safety are also addressed. Identifying knowledge gaps, our current effort will direct future research to advance potential applications of black cumin and TQ in health and diseases.
Collapse
Affiliation(s)
- Md. Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (A.A.M.S.); (M.T.H.); (M.T.-U.-A.)
| | - Md. Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Abdullah Al Mamun Sohag
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (A.A.M.S.); (M.T.H.); (M.T.-U.-A.)
| | - Md. Jamal Uddin
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (M.J.U.); (P.P.S.)
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Mahmudul Hasan Sikder
- Department of Pharmacology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Md. Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Gyeonggi-do, Anseong 17546, Korea;
| | - Binod Timalsina
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Partha Protim Sarker
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (M.J.U.); (P.P.S.)
- Department of Biotechnology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Mahboob Alam
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
- Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 780-714, Korea
| | - Md. Mohibbullah
- Department of Fishing and Post Harvest Technology, Sher-e-Bangla Agricultural University, Sher-e-Bangla Nagar, Dhaka 1207, Bangladesh;
| | - Md. Nazmul Haque
- Department of Fisheries Biology and Genetics, Patuakhali Science and Technology University, Patuakhali 8602, Bangladesh;
| | - Israt Jahan
- Department of Pharmacy, Faculty of Life and Earth Sciences, Jagannath University, Dhaka 1100, Bangladesh;
| | - Md. Tahmeed Hossain
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (A.A.M.S.); (M.T.H.); (M.T.-U.-A.)
| | - Tania Afrin
- Interdisciplinary Institute for Food Security, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Md. Mahbubur Rahman
- Research and Development Center, KNOTUS Co., Ltd., Yeounsu-gu, Incheon 22014, Korea;
| | - Md. Tahjib-Ul-Arif
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh; (A.A.M.S.); (M.T.H.); (M.T.-U.-A.)
| | - Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Diyah Fatimah Oktaviani
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Md Kawsar Khan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh;
- Department of Biological Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ho Jin Choi
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea; (M.A.H.); (R.D.); (B.T.); (Y.A.M.); (M.A.); (S.M.); (D.F.O.); (H.J.C.)
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
20
|
Almajali B, Al-Jamal HAN, Taib WRW, Ismail I, Johan MF, Doolaanea AA, Ibrahim WN. Thymoquinone, as a Novel Therapeutic Candidate of Cancers. Pharmaceuticals (Basel) 2021; 14:369. [PMID: 33923474 PMCID: PMC8074212 DOI: 10.3390/ph14040369] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
To date, natural products are widely used as pharmaceutical agents for many human diseases and cancers. One of the most popular natural products that have been studied for anticancer properties is thymoquinone (TQ). As a bioactive compound of Nigella sativa, TQ has shown anticancer activities through the inhibition of cell proliferation, migration, and invasion. The anticancer efficacy of TQ is being investigated in several human cancers such as pancreatic cancer, breast cancer, colon cancer, hepatic cancer, cervical cancer, and leukemia. Even though TQ induces apoptosis by regulating the expression of pro- apoptotic and anti-apoptotic genes in many cancers, the TQ effect mechanism on such cancers is not yet fully understood. Therefore, the present review has highlighted the TQ effect mechanisms on several signaling pathways and expression of tumor suppressor genes (TSG). Data from relevant published experimental articles on TQ from 2015 to June 2020 were selected by using Google Scholar and PubMed search engines. The present study investigated the effectiveness of TQ alone or in combination with other anticancer therapeutic agents, such as tyrosine kinase inhibitors on cancers, as a future anticancer therapy nominee by using nanotechnology.
Collapse
Affiliation(s)
- Belal Almajali
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia; (B.A.); (W.R.W.T.); (I.I.)
| | - Hamid Ali Nagi Al-Jamal
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia; (B.A.); (W.R.W.T.); (I.I.)
| | - Wan Rohani Wan Taib
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia; (B.A.); (W.R.W.T.); (I.I.)
| | - Imilia Ismail
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia; (B.A.); (W.R.W.T.); (I.I.)
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25594, Malaysia;
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health sciences, QU Health, Qatar University, Doha 2713, Qatar;
| |
Collapse
|
21
|
Omran Z, H. Dalhat M, Abdullah O, Kaleem M, Hosawi S, A Al-Abbasi F, Wu W, Choudhry H, Alhosin M. Targeting Post-Translational Modifications of the p73 Protein: A Promising Therapeutic Strategy for Tumors. Cancers (Basel) 2021; 13:1916. [PMID: 33921128 PMCID: PMC8071514 DOI: 10.3390/cancers13081916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 01/11/2023] Open
Abstract
The tumor suppressor p73 is a member of the p53 family and is expressed as different isoforms with opposing properties. The TAp73 isoforms act as tumor suppressors and have pro-apoptotic effects, whereas the ΔNp73 isoforms lack the N-terminus transactivation domain and behave as oncogenes. The TAp73 protein has a high degree of similarity with both p53 function and structure, and it induces the regulation of various genes involved in the cell cycle and apoptosis. Unlike those of the p53 gene, the mutations in the p73 gene are very rare in tumors. Cancer cells have developed several mechanisms to inhibit the activity and/or expression of p73, from the hypermethylation of its promoter to the modulation of the ratio between its pro- and anti-apoptotic isoforms. The p73 protein is also decorated by a panel of post-translational modifications, including phosphorylation, acetylation, ubiquitin proteasomal pathway modifications, and small ubiquitin-related modifier (SUMO)ylation, that regulate its transcriptional activity, subcellular localization, and stability. These modifications orchestrate the multiple anti-proliferative and pro-apoptotic functions of TAp73, thereby offering multiple promising candidates for targeted anti-cancer therapies. In this review, we summarize the current knowledge of the different pathways implicated in the regulation of TAp73 at the post-translational level. This review also highlights the growing importance of targeting the post-translational modifications of TAp73 as a promising antitumor strategy, regardless of p53 status.
Collapse
Affiliation(s)
- Ziad Omran
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (Z.O.); (O.A.)
| | - Mahmood H. Dalhat
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Omeima Abdullah
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (Z.O.); (O.A.)
| | - Mohammed Kaleem
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Salman Hosawi
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Fahd A Al-Abbasi
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, CA 94143, USA;
| | - Hani Choudhry
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Mahmoud Alhosin
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| |
Collapse
|
22
|
Ansary J, Giampieri F, Forbes-Hernandez TY, Regolo L, Quinzi D, Gracia Villar S, Garcia Villena E, Tutusaus Pifarre K, Alvarez-Suarez JM, Battino M, Cianciosi D. Nutritional Value and Preventive Role of Nigella sativa L. and Its Main Component Thymoquinone in Cancer: An Evidenced-Based Review of Preclinical and Clinical Studies. Molecules 2021; 26:molecules26082108. [PMID: 33916916 PMCID: PMC8067617 DOI: 10.3390/molecules26082108] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 11/16/2022] Open
Abstract
In recent times, scientific attention has been paid to different foods and their bioactive components for the ability to inhibit the onset and progress of different types of cancer. Nigella sativa extract, powder and seed oil and its main components, thymoquinone and α-hederin, have showed potent anticancer and chemosensitizing effects against various types of cancer, such as liver, colon, breast, renal, cervical, lung, ovarian, pancreatic, prostate and skin tumors, through the modulation of various molecular signaling pathways. Herein, the purpose of this review was to highlight the anticancer activity of Nigella sativa and it constitutes, focusing on different in vitro, in vivo and clinical studies and projects, in order to underline their antiproliferative, proapoptotic, cytotoxic and antimetastatic effects. Particular attention has been also given to the synergistic effect of Nigella sativa and it constitutes with chemotherapeutic drugs, and to the synthesized analogs of thymoquinone that seem to enhance the chemo-sensitizing potential. This review could be a useful step towards new research on N. sativa and cancer, to include this plant in the dietary treatments in support to conventional therapies, for the best achievement of therapeutic goals.
Collapse
Affiliation(s)
- Johura Ansary
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Francesca Giampieri
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tamara Y. Forbes-Hernandez
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Lucia Regolo
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Denise Quinzi
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Santos Gracia Villar
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Eduardo Garcia Villena
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
| | - Kilian Tutusaus Pifarre
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - José M. Alvarez-Suarez
- Departamento de Ingeniería en Alimentos, Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito, Quito 170157, Ecuador
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| | - Maurizio Battino
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| | - Danila Cianciosi
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| |
Collapse
|
23
|
Wu ZX, Yang Y, Zeng L, Patel H, Bo L, Lin L, Chen ZS. Establishment and Characterization of an Irinotecan-Resistant Human Colon Cancer Cell Line. Front Oncol 2021; 10:624954. [PMID: 33692943 PMCID: PMC7937870 DOI: 10.3389/fonc.2020.624954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. Irinotecan is widely used as a chemotherapeutic drug to treat CRC. However, the mechanisms of acquired resistance to irinotecan in CRC remain inconclusive. In the present study, we established a novel irinotecan-resistant human colon cell line to investigate the underlying mechanism(s) of irinotecan resistance, particularly the overexpression of ABC transporters. The irinotecan-resistant S1-IR20 cell line was established by exposing irinotecan to human S1 colon cancer cells. MTT cytotoxicity assay was carried out to determine the drug resistance profile of S1-IR20 cells. The drug-resistant cells showed about 47-fold resistance to irinotecan and cross-resistance to ABCG2 substrates in comparison with S1 cells. By Western blot analysis, S1-IR20 cells showed significant increase of ABCG2, but not ABCB1 or ABCC1 in protein expression level as compared to that of parental S1 cells. The immunofluorescence assay showed that the overexpressed ABCG2 transporter is localized on the cell membrane of S1-IR20 cells, suggesting an active efflux function of the ABCG2 transporter. This finding was further confirmed by reversal studies that inhibiting efflux function of ABCG2 was able to completely abolish drug resistance to irinotecan as well as other ABCG2 substrates in S1-IR20 cells. In conclusion, our work established an in vitro model of irinotecan resistance in CRC and suggested ABCG2 overexpression as one of the underlying mechanisms of acquired resistance to irinotecan. This novel resistant cell line may enable future studies to overcome drug resistance in vitro and improve CRC treatment in vivo.
Collapse
Affiliation(s)
- Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| | - Leli Zeng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States.,Precision Medicine Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| | - Lusheng Lin
- Cell Research Center, Shenzhen Bolun Institute of Biotechnology, Shenzhen, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| |
Collapse
|
24
|
Akter Z, Ahmed FR, Tania M, Khan MA. Targeting Inflammatory Mediators: An Anticancer Mechanism of Thymoquinone Action. Curr Med Chem 2021; 28:80-92. [PMID: 31604405 DOI: 10.2174/0929867326666191011143642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thymoquinone is a promising anticancer molecule, the chemopreventive role of which is well-known at least in vitro and in the animal model. In this review article, we focused on the anti-inflammatory activities of thymoquinone in cancer cells. METHOD Research data on inflammation, cancer and thymoquinone were acquired from PubMed, Scopus, Web of Science and Google Scholar. We reviewed papers published since the mid of the last century, and the most cited papers of the last ten years. RESULTS Studies indicate that thymoquinone possesses immunomodulatory activities, in addition to its chemopreventive role, as thymoquinone can target and modulate inflammatory molecules, like nuclear factor kappa B (NF-κβ), interleukins, tumor necrosis factor-α (TNF-α), and certain growth factors. As chronic inflammation plays an important role in cancer development, controlling inflammatory pathways is an important mechanism of an anticancer molecule, and modulation of inflammatory pathways might be one of the key mechanisms of thymoquinone's anticancer activities. CONCLUSION This article reviewed the role of inflammation on cancer development, and the action of thymoquinone on inflammatory molecules, which have been proved in vitro and in vivo. Much attention is required for studying the role of thymoquinone in immunotherapeutics and developing this molecule as a future anticancer drug.
Collapse
Affiliation(s)
- Zakia Akter
- Department of Biochemistry and Molecular Biology, Gono Bishwabidyalay, Dhaka, Bangladesh
| | - Faiza Rafa Ahmed
- Department of Biochemistry and Microbiology, North South University, Dhaka, Bangladesh
| | - Mousumi Tania
- Division of Molecular Cancer, Red Green Research Center, Dhaka, Bangladesh
| | - Md Asaduzzaman Khan
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Elgohary S, Elkhodiry AA, Amin NS, Stein U, El Tayebi HM. Thymoquinone: A Tie-Breaker in SARS-CoV2-Infected Cancer Patients? Cells 2021; 10:302. [PMID: 33540625 PMCID: PMC7912962 DOI: 10.3390/cells10020302] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Since the beginning of the SARS-CoV-2(severe acute respiratory syndrome-coronavirus-2) pandemic, arace to develop a vaccine has been initiated, considering the massive and rather significant economic and healthcare hits that this virus has caused. The pathophysiology occurring following COVID-19(coronavirus disease-2019) infection has givenhints regarding the supportive and symptomatic treatments to establish for patients, as no specific anti-SARS-CoV-2 is available yet. Patient symptoms vary greatly and range from mild symptoms to severe fatal complications. Supportive treatments include antipyretics, antiviral therapies, different combinations of broad-spectrum antibiotics, hydroxychloroquine and plasma transfusion. Unfortunately, cancer patients are at higher risk of viral infection and more likely to develop serious complications due to their immunocompromised state, the fact that they are already administering multiple medications, as well as combined comorbidity compared to the general population. It may seem impossible to find a drug that possesses both potent antiviral and anticancer effects specifically against COVID-19 infection and its complications and the existing malignancy, respectively. Thymoquinone (TQ) is the most pharmacologically active ingredient in Nigella sativa seeds (black seeds); it is reported to have anticancer, anti-inflammatory and antioxidant effects in various settings. In this review, we will discuss the multiple effects of TQ specifically against COVID-19, its beneficial effects against COVID-19 pathophysiology and multiple-organ complications, its use as an adjuvant for supportive COVID-19 therapy and cancer therapy, and finally, its anticancer effects.
Collapse
Affiliation(s)
- Sawsan Elgohary
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt; (S.E.); (A.A.E.); (N.S.A.)
| | - Aya A. Elkhodiry
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt; (S.E.); (A.A.E.); (N.S.A.)
| | - Nada S. Amin
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt; (S.E.); (A.A.E.); (N.S.A.)
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Hend M. El Tayebi
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt; (S.E.); (A.A.E.); (N.S.A.)
| |
Collapse
|
26
|
Almatroodi SA, Almatroudi A, Alsahli MA, Khan AA, Rahmani AH. Thymoquinone, an Active Compound of Nigella sativa: Role in Prevention and Treatment of Cancer. Curr Pharm Biotechnol 2020; 21:1028-1041. [DOI: 10.2174/1389201021666200416092743] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/30/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022]
Abstract
Background:
Cancer is the leading cause of death worldwide and the current mode of cancer
treatment causes side effects on normal cells and are still the key challenges in its’ treatment. However,
natural products or active compounds of medicinal plants have shown to be safe, affordable, and
effective in diseases cure.
Methods:
In this context, scientific studies evidence the health-promoting effects of natural products,
which work through its anti-oxidant, anti-inflammatory, and anti-cancer activity. Thymoquinone (TM),
a predominant active compound of Nigella sativa, has confirmed anti-neoplastic activity through its
ability to regulate various genetic pathways. In addition, thymoquinone has established anti-cancerous
effects through killing of various cancerous cells,and inhibiting the initiation, migration, invasion, and
progression of the cancer. The anti-cancer effects of TM are chiefly mediated via regulating various
cell signaling pathways such as VEGF, bcl2/bax ratio, p53, NF-kB, and oncogenes.
Results:
The anti-cancer drugs have limitations in efficacy and also causes adverse side effects on
normal cells. The combination of anti-cancer drugs and thymoquinone improves the efficacy of drugs
which is evident by decrease resistance to drugs and regulation of various cell signaling pathways.
Moreover, combination of anti-cancer drugs as well as thymoquinone shows synergistic effect on killing
of cancer cells and cells viability. Thus, TM, in combination with anti-cancer drugs, can be a good
strategy in the management of various types of cancer.
Conclusion:
In this review article, we deliver an outline of thymoquinone role in cancer inhibition and
prevention of cancer-based on in vivo and in vitro studies. Further studies on thymoquinone based on
clinical trials are highly required to explore the benefits of thymoquinone in cancer management.
Collapse
Affiliation(s)
- Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Mohammed A. Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Amjad A. Khan
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arshad H. Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
27
|
Ballout F, Monzer A, Fatfat M, Ouweini HE, Jaffa MA, Abdel-Samad R, Darwiche N, Abou-Kheir W, Gali-Muhtasib H. Thymoquinone induces apoptosis and DNA damage in 5-Fluorouracil-resistant colorectal cancer stem/progenitor cells. Oncotarget 2020; 11:2959-2972. [PMID: 32821342 PMCID: PMC7415406 DOI: 10.18632/oncotarget.27426] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
The high recurrence rates of colorectal cancer have been associated with a small population of cancer stem cells (CSCs) that are resistant to the standard chemotherapeutic drug, 5-fluorouracil (5FU). Thymoquinone (TQ) has shown promising antitumor properties on numerous cancer systems both in vitro and in vivo; however, its effect on colorectal CSCs is poorly established. Here, we investigated TQ's potential to target CSCs in a three-dimensional (3D) sphere-formation assay enriched for a population of colorectal cancer stem/progenitor cells. Our results showed a significant decrease in self-renewal potential of CSC populations enriched from 5FU-sensitive and resistant HCT116 cells at 10-fold lower concentrations when compared to 2D monolayers. TQ decreased the expression levels of colorectal stem cell markers CD44 and Epithelial Cell Adhesion Molecule EpCAM and proliferation marker Ki67 in colonospheres derived from both cell lines and reduced cellular migration and invasion. Further investigation revealed that TQ treatment led to increased TUNEL positivity and a dramatic increase in the amount of the DNA damage marker gamma H2AX particularly in 5FU-resistant colonospheres, suggesting that the diminished sphere forming ability in TQ-treated colonospheres is due to induction of DNA damage and apoptotic cell death. The intraperitoneal injection of TQ in mice inhibited tumor growth of spheres derived from 5FU-sensitive and 5FU-resistant HCT116 cells. Furthermore, TQ induced apoptosis and inhibited NF-κB and MEK signaling in mouse tumors. Altogether, our findings document TQ's effect on colorectal cancer stem-like cells and provide insights into its underlying mechanism of action.
Collapse
Affiliation(s)
- Farah Ballout
- Department of Biology, American University of Beirut, Lebanon
| | - Alissar Monzer
- Department of Biology, American University of Beirut, Lebanon
| | - Maamoun Fatfat
- Department of Biology, American University of Beirut, Lebanon
| | - Hala El Ouweini
- Department of Biology, American University of Beirut, Lebanon
| | - Miran A. Jaffa
- Department of Epidemiology and Population Health, American University of Beirut, Lebanon
| | - Rana Abdel-Samad
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Lebanon
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Lebanon
| | - Wassim Abou-Kheir
- Center for Drug Discovery and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Lebanon
| | - Hala Gali-Muhtasib
- Department of Biology, American University of Beirut, Lebanon
- Center for Drug Discovery and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Lebanon
| |
Collapse
|
28
|
Antioxidant, Anti-Inflammatory, and Microbial-Modulating Activities of Essential Oils: Implications in Colonic Pathophysiology. Int J Mol Sci 2020; 21:ijms21114152. [PMID: 32532055 PMCID: PMC7313461 DOI: 10.3390/ijms21114152] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Essential oils (EOs) are a complex mixture of hydrophobic and volatile compounds synthesized from aromatic plants, most of them commonly used in the human diet. In recent years, many studies have analyzed their antimicrobial, antioxidant, anti-inflammatory, immunomodulatory and anticancer properties in vitro and on experimentally induced animal models of colitis and colorectal cancer. However, there are still few clinical studies aimed to understand their role in the modulation of the intestinal pathophysiology. Many EOs and some of their molecules have demonstrated their efficacy in inhibiting bacterial, fungi and virus replication and in modulating the inflammatory and oxidative processes that take place in experimental colitis. In addition to this, their antitumor activity against colorectal cancer models makes them extremely interesting compounds for the modulation of the pathophysiology of the large bowel. The characterization of these EOs is made difficult by their complexity and by the different compositions present in the same oil having different geographical origins. This review tries to shift the focus from the EOs to their individual compounds, to expand their possible applications in modulating colon pathophysiology.
Collapse
|
29
|
Chemoresistance-Associated Silencing of miR-4454 Promotes Colorectal Cancer Aggression through the GNL3L and NF-κB Pathway. Cancers (Basel) 2020; 12:cancers12051231. [PMID: 32422901 PMCID: PMC7281507 DOI: 10.3390/cancers12051231] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/13/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
Guanine nucleotide-binding protein-like-3-like (GNL3L) is a crucial regulator of NF-κB signaling that is aberrantly activated during diverse chemoresistance-associated cellular processes. However, the molecular mechanisms of GNL3L tumor initiation and resistant state are largely unknown. Moreover, the identification of predictive biomarkers is necessary to effectively generate therapeutic strategies for metastatic human colorectal cancer (CRC). This study aims to identify how cells acquire resistance to anticancer drugs and whether the downregulation of miR-4454 is associated with the progression of CRC. Here, we have shown that the overexpression of miR-4454 in resistant tumors is a crucial precursor for the posttranscriptional repression of GNL3L in human chemoresistant CRC progression, and we used doxycycline induced miR-4454 overexpression that significantly reduced tumor volume in a subcutaneous injection nude mice model. Together, these observations highlight that the downregulation of miR-4454 in resistant clones is prominently responsible for maintaining their resistance against anticancer drug therapy. Our study indicates that the development of miR-4454 as a microRNA-based therapeutic approach to silence GNL3L may remarkably reduce oncogenic cell survival that depends on GNL3L/NF-κB signaling, making miR-4454 a candidate for treating metastatic human CRC.
Collapse
|
30
|
Ye SB, Cheng YK, Deng R, Deng Y, Li P, Zhang L, Lan P. The Predictive Value of Estrogen Receptor 1 on Adjuvant Chemotherapy in Locally Advanced Colorectal Cancer: A Retrospective Analysis With Independent Validation and Its Potential Mechanism. Front Oncol 2020; 10:214. [PMID: 32266127 PMCID: PMC7100261 DOI: 10.3389/fonc.2020.00214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/07/2020] [Indexed: 01/16/2023] Open
Abstract
Purpose: To investigate the predictive biomarker value of estrogen receptor 1 (ESR1) expression in tumor tissue on adjuvant chemotherapy in curatively resected colorectal cancer (CRC). Methods: A total of 467 CRC patients in 2007-2010 were retrospectively evaluated. Clinical information and follow-up data were retrieved from hospital registries and patient files. What's more, we used an external independent cohort (n = 511) from GSE39582 for further validation. Overall survival was estimated by the Kaplan-Meier method, and the survival curves were compared by log-rank tests. Cox proportional hazards models were used for multivariate analyses to calculate the hazard ratios (HRs) and test independent significance. Immunohistochemistry and Western blot were applied to detect protein expression of ESR1 in CRC patients and cell lines. The stable knockdown and overexpressed cells were transduced with the lentivirus. Cell viability was measured by an MTS reagent. Results: The predictive value of ESR1 was investigated in locally advanced CRC patients. Kaplan-Meier analysis indicated that ESR1 expression was significantly correlated with OS in patients receiving adjuvant chemotherapy from these cohorts, with p = 0.015 and p < 0.001, respectively. ESR1 expression was significantly correlated with 5-flurouracil (5-FU)-based adjuvant chemotherapy in training with an HR of 1.792 (95%CI: 1.100-2.921, p = 0.019). Downregulation of ESR1 was related with enhanced chemosensitivity to 5-FU in CRC cell lines, while upregulation of ESR1 was correlated with decreased chemosensitivity. Conclusions: The present study manifest clinical validity of ESR1 expression as a predictive biomarker on 5-FU-based adjuvant chemotherapy in stage II-III CRC.
Collapse
Affiliation(s)
- Shu-Biao Ye
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Kan Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Radiation Oncology, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Ru Deng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Oncology, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Deng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Oncology, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peisi Li
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Nordin A, Kamal H, Yazid MD, Saim A, Idrus R. Effect of Nigella sativa and its bioactive compound on type 2 epithelial to mesenchymal transition: a systematic review. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:290. [PMID: 31666058 PMCID: PMC6821016 DOI: 10.1186/s12906-019-2706-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nigella sativa or commonly known as black seed or black cumin is one of the most ubiquitous complementary medicine. Epithelial to mesenchymal transition (EMT) of type 2 is defined by the balance between wound healing and tissue fibrosis, which is dependent to the state of inflammation. This systematic review is conducted to provide an overview regarding the reported effect of Nigella sativa and its bioactive compound on the type 2 EMT. METHODS A search was done in EBSCOHOST, OVID and SCOPUS database to obtain potentially relevant articles that were published between 1823 and August 2019. This review includes studies that focus on the effect of Nigella sativa and its bioactive compound on the events related to type 2 EMT. RESULTS A total of 1393 research articles were found to be potentially related to the effect of Nigella sativa and its bioactive compound, thymoquinone on Type 2 EMT. After screening was done, 22 research articles met inclusion criteria and were included in this review. Majority of the studies, reported better wound healing rate or significant prevention of tissue inflammation and organ fibrosis following Nigella sativa or thymoquinone treatments. In terms of wound healing, studies included reported progression of EMT related pathological changes after treatment with Nigella sativa or thymoquinone. Alternatively, in terms of fibrosis and inflammation, studies included reported reversal of pathological changes related to EMT after treatment with Nigella sativa or thymoquinone. CONCLUSION Through this review, Nigella sativa and thymoquinone have been associated with events in Type 2 EMT. They have been shown to promote wound healing, attenuate tissue inflammation, and prevent organ fibrosis via regulation of the EMT process.
Collapse
Affiliation(s)
- Abid Nordin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur Malaysia
- Nordin Kamil Consulting, 30-2, Jalan Dwitasik, Dataran Dwitasik, 56000 Cheras, Kuala Lumpur Malaysia
| | - Haziq Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur Malaysia
| | - Aminuddin Saim
- Ear, Nose & Throat Consultant Clinic, Ampang Puteri Specialist Hospital, 68000 Ampang, Selangor Malaysia
| | - Ruszymah Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur Malaysia
| |
Collapse
|
32
|
Thymoquinone (2-Isoprpyl-5-methyl-1, 4-benzoquinone) as a chemopreventive/anticancer agent: Chemistry and biological effects. Saudi Pharm J 2019; 27:1113-1126. [PMID: 31885471 PMCID: PMC6921197 DOI: 10.1016/j.jsps.2019.09.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer remains the topmost disorders of the mankind and number of cases is unceasingly growing at unprecedented rates. Although the synthetic anti-cancer compounds still hold the largest market in the modern treatment of cancer, natural agents have always been tried and tested for potential anti-cancer properties. Thymoquinone (TQ), a monoterpene and main ingredient in the essential oil of Nigella sativa L. has got very eminent rankings in the traditional systems of medicine for its anti-cancer pharmacological properties. In this review we summarized the diverse aspects of TQ including its chemistry, biosynthesis, sources and pharmacological properties with a major concern being attributed to its anti-cancer efficacies. The role of TQ in different aspects involved in the pathogenesis of cancer like inflammation, angiogenesis, apoptosis, cell cycle regulation, proliferation, invasion and migration have been described. The mechanism of action of TQ in different cancer types has been briefly accounted. Other safety and toxicological aspects and some combination therapies involving TQ have also been touched. A detailed literature search was carried out using various online search engines like google scholar and pubmed regarding the available research and review accounts on thymoquinone upto may 2019. All the articles reporting significant addition to the activities of thymoquinone were selected. Additional information was acquired from ethno botanical literature focusing on thymoquinone. The compound has been the centre of attention for a long time period and researched regularly in quite considerable numbers for its various physicochemical, medicinal, biological and pharmacological perspectives. Thymoquinone is studied for various chemical and pharmacological activities and demonstrated promising anti-cancer potential. The reviewed reports confirmed the strong anti-cancer efficacy of thymoquinone. Further in-vitro and in-vivo research is strongly warranted regarding the complete exploration of thymoquinone in ethnopharmacological context.
Collapse
Key Words
- AMPK, AMP-activated protein kinase
- APC, adenomatous polyposis coli
- Anti-cancer therapeutics
- CDDP, cisplatin
- CDKs, cyclin-dependent kinases
- EMT, epithelial to mesenchymal transition
- FGFs, fibroblast growth factors
- FTIR, fourier-transform infrared spectroscopy
- GBM, glioblastoma multiforme
- HPDE, human pancreatic ductal epithelial cells
- IUPAC, international union of pure and applied chemistry
- LKB1, liver kinase B1
- LPS, lipopolysaccharide
- MC-A, myrtucommulone-A
- NLCs, nanostructured lipid carriers
- NMR, nuclear magnetic resonance
- NSAIDs, non-steroidal anti-inflammatory drugs
- Natural compounds
- OEC, oral epithelial cells
- PCNA, proliferating cell nuclear antigen
- PXRD, powder x-ray diffraction
- Phytopharmaceuticals
- Plant products
- RES, resveratrol
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SCLC, small cell lung carcinoma
- SLNs, solid lipid nanoparticles
- THQ, thymohydroquinone
- TMZ, temozolomide
- TNBC, triple negative breast cancer
- TNFα, tumor necrosis factor alpha
- TQ, thymoquinone
- Thymoquinone
- UMSCC, university of Michigan squamous cell carcinoma
- USD, United States Dollar
- VEGF, vascular endothelial growth factor
- WHO, world health organization
- XIAP, X-linked inhibitor of apoptosis protein
- eEF-2K, elongation factor 2 kinase
Collapse
|
33
|
Pezzani R, Salehi B, Vitalini S, Iriti M, Zuñiga FA, Sharifi-Rad J, Martorell M, Martins N. Synergistic Effects of Plant Derivatives and Conventional Chemotherapeutic Agents: An Update on the Cancer Perspective. ACTA ACUST UNITED AC 2019; 55:medicina55040110. [PMID: 30999703 PMCID: PMC6524059 DOI: 10.3390/medicina55040110] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 03/28/2019] [Accepted: 04/10/2019] [Indexed: 01/07/2023]
Abstract
Synergy is a process in which some substances cooperate to reach a combined effect that is greater than the sum of their separate effects. It can be considered a natural "straight" strategy which has evolved by nature to obtain more efficacy at low cost. In this regard, synergistic effects may be observed in the interaction between herbal products and conventional drugs or biochemical compounds. It is important to identify and exploit these interactions since any improvement brought by such kind of process can be advantageously used to treat human disorders. Even in a complex disease such as cancer, positive synergistic plant-drug interactions should be investigated to achieve the best outcomes, including providing a greater benefit to patients or avoiding adverse side effects. This review analyzes and summarizes the current knowledge on the synergistic effects of plant-drug interactions with a focus on anticancer strategies.
Collapse
Affiliation(s)
- Raffaele Pezzani
- Endocrinology Unit, Department of Medicine, University of Padova, via Ospedale 105, 35128 Padova, Italy.
- AIROB-Associazione Italiana per la Ricerca Oncologica di Base, 3520128 Padova, Italy.
| | - Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam 44340847, Iran.
| | - Sara Vitalini
- Department of Agricultural and Environmental Sciences, Milan State University, via G. Celoria 2, 20133 Milan, Italy.
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences, Milan State University, via G. Celoria 2, 20133 Milan, Italy.
| | - Felipe Andrés Zuñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepcion 4070386, Chile.
| | - Javad Sharifi-Rad
- Food Safety Research Center (Salt), Semnan University of Medical Sciences, Semnan 3519899951, Iran.
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepcion 4070386, Chile.
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
34
|
Kirsanov KI, Vlasova OA, Fetisov TI, Zenkov RG, Lesovaya EA, Belitsky GA, Gurova K, Yakubovskaya MG. Influence of DNA-binding compounds with cancer preventive activity on the mechanisms of gene expression regulation. ADVANCES IN MOLECULAR ONCOLOGY 2019. [DOI: 10.17650/2313-805x-2018-5-4-41-63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- K. I. Kirsanov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia; Peoples’ Friendship University of Russia
| | - O. A. Vlasova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - T. I. Fetisov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - R. G. Zenkov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - E. A. Lesovaya
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia; I.P. Pavlov Ryazan State Medical University
| | - G. A. Belitsky
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | | | - M. G. Yakubovskaya
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| |
Collapse
|
35
|
Zhang Y, Fan Y, Huang S, Wang G, Han R, Lei F, Luo A, Jing X, Zhao L, Gu S, Zhao X. Thymoquinone inhibits the metastasis of renal cell cancer cells by inducing autophagy via AMPK/mTOR signaling pathway. Cancer Sci 2018; 109:3865-3873. [PMID: 30259603 PMCID: PMC6272120 DOI: 10.1111/cas.13808] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/19/2022] Open
Abstract
Thymoquinone (TQ, 2-methyl-5-isopropyl-1,4-benzoquinone), a bioactive constituent extracted from the seeds of Nigella sativa, has been proved to exert anti-tumor efficiency in various cancers. Autophagy is a self-digestion phenomenon, and its role in tumor formation and progression remains controversial. In the present study, we investigated the effects of TQ on renal cell cancer (RCC) cell lines (786-O and ACHN) using wound healing assay, transwell assay and western blot analysis. We found that TQ effectively inhibited the metastatic capacity of RCC cells in vitro, which was also verified in a xenograft model. Meanwhile, we observed LC3 puncta and detected the expression of LC3 in TQ-treated RCC cells, and then found that autophagy was induced by TQ in 786-O and ACHN cell lines. In addition, TQ inhibited the migration and invasion as well as the EMT in RCC cells in an autophagy-dependent manner. To further explore the underlying mechanism, we detected the AMPK/mTOR signaling pathway. The results indicated that TQ inhibited the metastasis of RCC cells by inducing autophagy via AMPK/mTOR signaling pathway. In conclusion, our findings provide a novel therapeutic strategy that aims at TQ-induced autophagy in RCC treatment.
Collapse
Affiliation(s)
- Yujiao Zhang
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yizeng Fan
- Department of Urologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Shangke Huang
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Guanying Wang
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Rui Han
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Fuxi Lei
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Anqi Luo
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xin Jing
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Lin Zhao
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Shanzhi Gu
- Department of College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina
| | - Xinhan Zhao
- Department of Medical Oncologythe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
36
|
Imran M, Rauf A, Khan IA, Shahbaz M, Qaisrani TB, Fatmawati S, Abu-Izneid T, Imran A, Rahman KU, Gondal TA. Thymoquinone: A novel strategy to combat cancer: A review. Biomed Pharmacother 2018; 106:390-402. [PMID: 29966985 DOI: 10.1016/j.biopha.2018.06.159] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
The higher consumption of fruit, herbs, spices, and vegetables is well known and practical strategy to cure human cancers owing to their presence of bioactive compounds. Among these, Nigella sativa is a promising source of bioactive compounds including thymoquinone, monoterpenes, p-cymene and α-piene etc. Thymoquinone has been found effective to inhibit the different cancer stages such as proliferation, migration and invasion. It also acts as anticancer agent against different human cancers such as breast, pancreatic, prostate, blood, oral, bone, head and neck, cervical, liver and lung. It significantly mediated miR-34a up-regulation, enhanced the levels of miR-34a through p53, and down controlled Rac1 expression. Thymoquinone induces apoptosis, regulates the levels of pro- and anti- apoptotic genes. It also has been known to lower the phosphorylation of NF-κB and IKKα/β and reduces the metastasis as well as also lowered the ERK1/2 and PI3K activities. Thymoquinone inhibits the metastasis through activation of JNK and p38. The present review article highlights the anticancer perspectives of thymoquinone in human by various pathways and use of this compound as diet based therapy has proven new pharmacological agent against several types of cancers.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan.
| | - Imtiaz Ali Khan
- Department ofAgriculture, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Shahbaz
- Department of Food science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | | | - Sri Fatmawati
- Department of Chemistry,Faculty of Mathematics and Natural Sciences, Institut Teknologi Sepuluh Nopember, Kampus ITS-Sukolilo, Surabaya, Indonesia
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, P.O.Box 42, Saudi Arabia
| | - Ali Imran
- Institute of Home and Food Sciences, Faculty of Science and Technology, Government College University, Faisalabad, Pakistan
| | - Khaliq Ur Rahman
- Department of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Centre of Advanced Sensory Science, Deakin University, Australia
| |
Collapse
|
37
|
Huang RZ, Jin L, Wang CG, Xu XJ, Du Y, Liao N, Ji M, Liao ZX, Wang HS. A pentacyclic triterpene derivative possessing polyhydroxyl ring A suppresses growth of HeLa cells by reactive oxygen species-dependent NF-κB pathway. Eur J Pharmacol 2018; 838:157-169. [PMID: 30153443 DOI: 10.1016/j.ejphar.2018.08.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Pentacyclic triterpene derivatives possessing polyhydroxyl ring A exhibit many important pharmacological activities. (1β, 2α, 3β, 19β, 23)-1,2,3,19,23-pentahydroxyolean-12-en-28-oic acid (5), a new bioactive phytochemical with tetra-hydroxyl ring A isolated from Euphorbia sieboldiana in our laboratory, showed potential inhibition effects against several cancer cells previously. This study was performed to investigate the underlying mechanisms of action for its antitumor activity. The results showed that compound 5 inhibited dose-/time-dependently cell growth with low toxicity to normal cells and induced apoptosis in cervical cancer cells. Also, compound 5 inhibited the growth and proliferation of HeLa cells and resulted in G1 phase arrest. Furthermore, exposure of cells to compound 5 caused inactivation of the TNF-α-TAK1-IKK-NF-κB axis and inhibition of TNF-α-stimulated NF-κB activity, followed by down-regulation of NF-κB target genes involved in cell apoptosis (Bcl-2) and in the cell cycle and growth (Cyclin D, c-Myc). Additionally, compound 5 significantly suppressed the migration of HeLa cells. In addition, exposure of HeLa cells to compound 5 decreased the activity of NF-κB through the generation of reactive oxygen species (ROS). Collectively, these results suggested that compound 5 exerted potent anticancer effects on HeLa cells in vitro through targeting the ROS-dependent NF-κB signaling cascade and this compound may be a promising anticancer agent for cancer treatment.
Collapse
Affiliation(s)
- Ri-Zhen Huang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Le Jin
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chun-Gu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, PR China
| | - Xiao-Jing Xu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ying Du
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Na Liao
- Department of Pharmacy, College of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhi-Xin Liao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Heng-Shan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
38
|
Chen HP, Lee YK, Huang SY, Shi PC, Hsu PC, Chang CF. Phthalate exposure promotes chemotherapeutic drug resistance in colon cancer cells. Oncotarget 2017; 9:13167-13180. [PMID: 29568348 PMCID: PMC5862569 DOI: 10.18632/oncotarget.23481] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/08/2017] [Indexed: 12/30/2022] Open
Abstract
Phthalates are widely used as plasticizers. Humans can be exposed to phthalates through ingestion, inhalation, or treatments that release di(2-ethylhexyl) phthalate (DEHP) and its metabolite, mono(2-ehylhexyl) phthalate (MEHP), into the body from polyvinyl chloride-based medical devices. Phthalate exposure may induce reproductive toxicity, liver damage, and carcinogenesis in humans. This study found that colon cancer cells exposed to DEHP or MEHP exhibited increased cell viability and increased levels of P-glycoprotein, CD133, Bcl-2, Akt, ERK, GSK3β, and β-catenin when treated with oxaliplatin or irinotecan, as compared to control. The P-glycoprotein inhibitor, tariquidar, which blocks drug efflux, reduced the viability of DEHP- or MEHP-treated, anti-cancer drug-challenged cells. DEHP or MEHP treatment also induced colon cancer cell migration and epithelial-mesenchymal transformation. Elevated stemness-related protein levels (β-catenin, Oct4, Sox2, and Nanog) and increased cell sphere sizes indicated that DEHP- or MEHP-treated cells were capable of self-renewal. We also found that serum DEHP concentrations were positively correlated with cancer recurrence. These results suggest phthalate exposure enhances colon cancer cell metastasis and chemotherapeutic drug resistance by increasing cancer cell stemness, and that P-glycoprotein inhibitors might improve outcomes for advanced or drug-resistant colon cancer patients.
Collapse
Affiliation(s)
- Hsin-Pao Chen
- Department of Surgery, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan.,Department of Safety, Health and Environmental Engineering, National Kaohsiung First University of Science and Technology, Kaohsiung 811, Taiwan
| | - Yung-Kuo Lee
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shih Yin Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pei-Chun Shi
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ping-Chi Hsu
- Department of Safety, Health and Environmental Engineering, National Kaohsiung First University of Science and Technology, Kaohsiung 811, Taiwan
| | - Chuan-Fa Chang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
39
|
Duan L, Deng L, Wang D, Ma S, Li C, Zhao D. Treatment mechanism of matrine in combination with irinotecan for colon cancer. Oncol Lett 2017; 14:2300-2304. [PMID: 28781667 PMCID: PMC5530135 DOI: 10.3892/ol.2017.6407] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/25/2017] [Indexed: 12/23/2022] Open
Abstract
The inhibitory effect of matrine (MA) was studied in combination with irinotecan (CPT-11) on proliferation of human colon carcinoma cell line HT29. We also explored the mechanism of cell apoptosis induction in HT29. HT29 cells were treated with different concentrations of MA and CPT-11 alone and in combination. The growth inhibition in HT29 cells was evaluated using MTT assay. Apoptosis was detected using AV-PI double staining flow cytometry. Transmission electron microscopy was used to detect structural changes in cells. Topoisomerase (TOPO) I, Bax and Caspase-3 expression levels were evaluated using western blot analysis. MA and CPT-11 alone and in combination, inhibited the proliferation of HT29 cells, whereas the combination treatment exhibited higher inhibitory effect (P<0.01). This suggests the existence of synergistic cytotoxicity. Compared with each treatment alone, the combination treatment caused more significant damage to cell structure, and caused a significantly higher apoptosis rate (P<0.01). Additionally, the combination treatment increased TOPO I, Bax and Caspase-3 expression levels (P<0.01). In conclusion, MA in combination with CPT-11 synergistically inhibited HT29 cell proliferation and induced apoptosis in these cells. The mechanism may be related to upregulation of the TOPO I, Bax and Caspase-3 protein expression.
Collapse
Affiliation(s)
- Ling Duan
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Leijiao Deng
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Dabin Wang
- Department of Anesthesiology, The People's Hospital of Gansu Province, Lanzhou, Gansu 730000, P.R. China
| | - Shoucheng Ma
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chunmei Li
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Da Zhao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
40
|
Fröhlich T, Ndreshkjana B, Muenzner JK, Reiter C, Hofmeister E, Mederer S, Fatfat M, El-Baba C, Gali-Muhtasib H, Schneider-Stock R, Tsogoeva SB. Synthesis of Novel Hybrids of Thymoquinone and Artemisinin with High Activity and Selectivity Against Colon Cancer. ChemMedChem 2017; 12:226-234. [PMID: 27973725 DOI: 10.1002/cmdc.201600594] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Indexed: 12/28/2022]
Abstract
Colorectal cancer causes 0.5 million deaths each year. To combat this type of cancer the development of new specific drug candidates is urgently needed. In the present work seven novel thymoquinone-artemisinin hybrids with different linkers were synthesized and tested for their in vitro anticancer activity against a panel of various tumor cell lines. The thymoquinone-artesunic acid hybrid 7 a, in which both subunits are connected via an ester bond, was found to be the most active compound and selectively decreased the viability of colorectal cancer cells with an IC50 value of 2.4 μm (HCT116) and 2.8 μm (HT29). Remarkably, hybrid 7 a was up to 20-fold more active than its parent compounds (thymoquinone and artesunic acid), while not affecting nonmalignant colon epithelial HCEC cells (IC50 >100 μm). Moreover, the activity of hybrid 7 a was superior to that of various 1:1 mixtures of thymoquinone and artesunic acid. Furthermore, hybrid 7 a was even more potent against both colon cancer cell lines than the clinically used drug 5-fluorouracil. These results are another excellent proof of the hybridization concept and confirm that the type and length of the linker play a crucial role for the biological activity of a hybrid drug. Besides an increase in reactive oxygen species (ROS), elevated levels of the DNA-damage marker γ-H2AX were observed. Both effects seem to be involved in the molecular mechanism of action for hybrid 7 a in colorectal cancer cells.
Collapse
Affiliation(s)
- Tony Fröhlich
- Organic Chemistry Chair I and Interdisciplinary Center for Molecular Materials (ICMM), Friedrich Alexander University of Erlangen-Nürnberg, Henkestr. 42, 91054, Erlangen, Germany
| | - Benardina Ndreshkjana
- Experimental Tumor Pathology, Institute of Pathology, Friedrich Alexander University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany
| | - Julienne K Muenzner
- Experimental Tumor Pathology, Institute of Pathology, Friedrich Alexander University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany
| | - Christoph Reiter
- Organic Chemistry Chair I and Interdisciplinary Center for Molecular Materials (ICMM), Friedrich Alexander University of Erlangen-Nürnberg, Henkestr. 42, 91054, Erlangen, Germany
| | - Elisabeth Hofmeister
- Organic Chemistry Chair I and Interdisciplinary Center for Molecular Materials (ICMM), Friedrich Alexander University of Erlangen-Nürnberg, Henkestr. 42, 91054, Erlangen, Germany
| | - Sandra Mederer
- Experimental Tumor Pathology, Institute of Pathology, Friedrich Alexander University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany
| | - Maamoun Fatfat
- Department of Biology, Department of Anatomy, Cell Biology and Physiological Science, American University of Beirut, Beirut, Lebanon
| | - Chirine El-Baba
- Experimental Tumor Pathology, Institute of Pathology, Friedrich Alexander University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany
| | - Hala Gali-Muhtasib
- Department of Biology, Department of Anatomy, Cell Biology and Physiological Science, American University of Beirut, Beirut, Lebanon
| | - Regine Schneider-Stock
- Experimental Tumor Pathology, Institute of Pathology, Friedrich Alexander University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany
| | - Svetlana B Tsogoeva
- Organic Chemistry Chair I and Interdisciplinary Center for Molecular Materials (ICMM), Friedrich Alexander University of Erlangen-Nürnberg, Henkestr. 42, 91054, Erlangen, Germany
| |
Collapse
|
41
|
Gutova M, Goldstein L, Metz M, Hovsepyan A, Tsurkan LG, Tirughana R, Tsaturyan L, Annala AJ, Synold TW, Wan Z, Seeger R, Anderson C, Moats RA, Potter PM, Aboody KS. Optimization of a Neural Stem-Cell-Mediated Carboxylesterase/Irinotecan Gene Therapy for Metastatic Neuroblastoma. MOLECULAR THERAPY-ONCOLYTICS 2016; 4:67-76. [PMID: 28345025 PMCID: PMC5363723 DOI: 10.1016/j.omto.2016.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022]
Abstract
Despite improved survival for children with newly diagnosed neuroblastoma (NB), recurrent disease is a significant problem, with treatment options limited by anti-tumor efficacy, patient drug tolerance, and cumulative toxicity. We previously demonstrated that neural stem cells (NSCs) expressing a modified rabbit carboxylesterase (rCE) can distribute to metastatic NB tumor foci in multiple organs in mice and convert the prodrug irinotecan (CPT-11) to the 1,000-fold more toxic topoisomerase-1 inhibitor SN-38, resulting in significant therapeutic efficacy. We sought to extend these studies by using a clinically relevant NSC line expressing a modified human CE (hCE1m6-NSCs) to establish proof of concept and identify an intravenous dose and treatment schedule that gave maximal efficacy. Human-derived NB cell lines were significantly more sensitive to treatment with hCE1m6-NSCs and irinotecan as compared with drug alone. This was supported by pharmacokinetic studies in subcutaneous NB mouse models demonstrating tumor-specific conversion of irinotecan to SN-38. Furthermore, NB-bearing mice that received repeat treatment with intravenous hCE1m6-NSCs and irinotecan showed significantly lower tumor burden (1.4-fold, p = 0.0093) and increased long-term survival compared with mice treated with drug alone. These studies support the continued development of NSC-mediated gene therapy for improved clinical outcome in NB patients.
Collapse
Affiliation(s)
- Margarita Gutova
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Leanne Goldstein
- Information Sciences, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Marianne Metz
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Anahit Hovsepyan
- Departments of Radiology and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Lyudmila G Tsurkan
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38101, USA
| | - Revathiswari Tirughana
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lusine Tsaturyan
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Alexander J Annala
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Timothy W Synold
- Department of Cancer Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Zesheng Wan
- Children's Center for Cancer and Blood Diseases, CHLA/Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Robert Seeger
- Children's Center for Cancer and Blood Diseases, CHLA/Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Clarke Anderson
- Department of Pediatric Oncology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Rex A Moats
- Departments of Radiology and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Philip M Potter
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38101, USA
| | - Karen S Aboody
- Departments of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|