1
|
Famurewa AC, Akhigbe RE, George MY, Adekunle YA, Oyedokun PA, Akhigbe TM, Fatokun AA. Mechanisms of ferroptotic and non-ferroptotic organ toxicity of chemotherapy: protective and therapeutic effects of ginger, 6-gingerol and zingerone in preclinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4747-4778. [PMID: 39636404 PMCID: PMC11985630 DOI: 10.1007/s00210-024-03623-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Chemotherapy (CT) is one of the flagship options for the treatment of cancers worldwide. It involves the use of cytotoxic anticancer agents to kill or inhibit the proliferation of cancer cells. However, despite its clinical efficacy, CT triggers side effect toxicities in several organs, which may impact cancer patient's quality of life and treatment outcomes. While the side effect toxicity is consistent with non-ferroptotic mechanisms involving oxidative stress, inflammation, mitochondrial impairment and other aberrant signalling leading to apoptosis and necroptosis, recent studies show that ferroptosis, a non-apoptotic, iron-dependent cell death pathway, is also involved in the pathophysiology of CT organ toxicity. CT provokes organ ferroptosis via system Xc-/GPX-4/GSH/SLC7A11 axis depletion, ferritinophagy, iron overload, lipid peroxidation and upregulation of ferritin-related proteins. Cisplatin (CP) and doxorubicin (DOX) are common CT drugs indicated to induce ferroptosis in vitro and in vivo. Studies have explored natural preventive and therapeutic strategies using ginger rhizome and its major bioactive compounds, 6-gingerol (6G) and zingerone (ZG), to combat mechanisms of CT side effect toxicity. Ginger extract, 6G and ZG mitigate non-ferroptotic oxidative inflammation, apoptosis and mitochondrial dysfunction mechanisms of CT side effect toxicity, but their effects on CT-induced ferroptosis remain unclear. Systematic investigations are, therefore, needed to unfold the roles of ginger, 6G and ZG on ferroptosis involved in CT side effect toxicity, as they are potential natural agents for the prevention of CT toxicity. This review reveals the ferroptotic and non-ferroptotic toxicity mechanisms of CT and the protective mechanisms of ginger, 6G and ZG against CT-induced, ferroptotic and non-ferroptotic organ toxicities.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike, Ikwo, Nigeria.
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Roland E Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Yemi A Adekunle
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Precious A Oyedokun
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Tunmise M Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Breeding and Genetics Unit, Department of Agronomy, Osun State University, Osogbo, Osun State, Nigeria
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
2
|
Sravani A, Thomas J. Targeting epithelial-mesenchymal transition signaling pathways with Dietary Phytocompounds and repurposed drug combinations for overcoming drug resistance in various cancers. Heliyon 2025; 11:e41964. [PMID: 39959483 PMCID: PMC11830326 DOI: 10.1016/j.heliyon.2025.e41964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a crucial step in metastasis formation. It enhances the ability of cancer cells' to self-renew and initiate tumors, while also increasing resistance to apoptosis and chemotherapy. Among the signaling pathways a few signaling pathways such as Notch, TGF-beta, and Wnt-beta catenin are critically involved in the epithelial-to-mesenchymal transition (EMT) acquisition. Therefore, regulating EMT is a key strategy for controlling malignant cell behavior. This is done by interconnecting other signaling pathways in many cancer types. Although there is extensive preclinical evidence regarding EMT's function in the development of cancer, there is still a deficiency in clinical translation at the therapeutic level. Thus, there is a need for medications that are both highly effective and with low cytotoxic for modulating EMT transitions at ground level. Thus, this led to the study of the evaluation and efficiency of phytochemicals found in dietary sources of fruits and vegetables and also the combination of small molecular repurposed drugs that can enhance the effectiveness of traditional cancer treatments. This review summarises major EMT-associated pathways and their cross talks with their mechanistic insights and the role of different dietary phytochemicals (curcumin, ginger, fennel, black pepper, and clove) and their natural analogs and also repurposed drugs (metformin, statin, chloroquine, and vitamin D) which are commonly used in regulating EMT in various preclinical studies. This review also investigates the concept of low-toxicity and broad spectrum ("The Halifax Project") approach which can help for site targeting of several key pathways and their mechanism. We also discuss the mechanisms of action, models for our dietary phytochemicals, and repurposed drugs and their combinations used to identify potential anti-EMT activities. Additionally, we also analyzed existing literature and proposed new directions for accelerating the discovery of novel drug candidates that are safe to administer.
Collapse
Affiliation(s)
- A.N.K.V. Sravani
- Center for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - John Thomas
- Center for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
3
|
Adikesavan M, Athiraja P, Divakar MBB. Investigation on the Anticancer Activity of [6]-Gingerol of Zingiber officinale and its Structural Analogs against Skin Cancer. Curr Comput Aided Drug Des 2024; 20:367-373. [PMID: 37076965 DOI: 10.2174/1573409919666230418095105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/20/2023] [Accepted: 02/20/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION Skin cancer is the most common type of cancer caused by the uncontrolled growth of abnormal cells in the epidermis and the outermost skin layer. AIM This study aimed to study the anti-skin cancer potential of [6]-Gingerol and 21 related structural analogs using in vitro and in silico studies. METHODS The ethanolic crude extract of the selected plant was subjected to phytochemical and GC-MS analysis to confirm the presence of the [6]-gingerol. The anticancer activity of the extract was evaluated by MTT (3-[4, 5-dimethylthiazol-2-y]-2, 5-diphenyl tetrazolium bromide) assay using the A431 human skin adenocarcinoma cell line. RESULTS The GC-MS analysis confirmed the presence of [6]-Gingerol compound, and its promising cytotoxicity IC50 was found at 81.46 ug/ml in the MTT assay. Furthermore, the in silico studies used [6]-Gingerol and 21 structural analogs collected from the PubChem database to investigate the anticancer potential and drug-likeliness properties. Skin cancer protein, DDX3X, was selected as a target that regulates all stages of RNA metabolism. It was docked with 22 compounds, including [6]-Gingerol and 21 structural analogs. The potent lead molecule was selected based on the lowest binding energy value. CONCLUSION Thus, the [6]-Gingerol and its structure analogs could be used as lead molecules against skin cancer and future drug development process.
Collapse
Affiliation(s)
- Monisha Adikesavan
- Department of Biotechnology, Prathyusha Engineering College, Thiruvallur, 602025, Tamil Nadu, India
| | - Praveena Athiraja
- Department of Biotechnology, Prathyusha Engineering College, Thiruvallur, 602025, Tamil Nadu, India
| | | |
Collapse
|
4
|
Asghari Lalami Z, Tafvizi F, Naseh V, Salehipour M. Fabrication, optimization, and characterization of pH-responsive PEGylated nanoniosomes containing gingerol for enhanced treatment of breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3867-3886. [PMID: 37368028 DOI: 10.1007/s00210-023-02579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
Multiple potential drug delivery strategies have emerged as a result of recent advances in nanotechnology and nanomedicine. The aim of this research was to prepare an optimized system of PEGylated gingerol-loaded niosomes (Nio-Gin@PEG) as an excellent candidate for the treatment of human breast cancer cells. The preparation procedure was modified by adjusting the drug concentration, lipid content, and Span60/Tween60 ratio, resulting in high encapsulation efficacy (EE%), rapid release rate, and reduced size. The Nio-Gin@PEG exhibited significantly improved storage stability compared to the gingerol-loaded niosomes formulation (Nio-Gin), with minimal changes in EE%, release profile, and size during storage. Furthermore, Nio-Gin@PEG demonstrated pH-dependent release behavior, with delayed drug diffusion at physiological pH and significant drug diffusion under acidic conditions (pH = 5.4), making it a promising option for cancer treatment. Cytotoxicity tests indicated that Nio-Gin@PEG possessed excellent biocompatibility with human fibroblast cells while exerting a remarkable inhibitory effect on MCF-7 and SKBR3 breast cancer cells, attributed to the presence of gingerol and the PEGylated structure in the preparation. Nio-Gin@PEG also exhibited the ability to modulate the expression of target genes. We observed statistically significant down-regulation of the expression of BCL2, MMP2, MMP9, HER2, CCND1, CCNE1, BCL2, CDK4, and VEGF genes, along with up-regulation of the expression of BAX, CASP9, CASP3, and P21 genes. Flow cytometry results revealed that Nio-Gin@PEG could induce a higher rate of apoptosis in both cancerous cells compared to gingerol and Nio-Gin, owing to the optimal encapsulation and efficient drug release from the formulation, as confirmed by cell cycle tests. ROS generation demonstrated the superior antioxidant effect of Nio-Gin@PEG compared to other prepared formulations. The results of this study emphasize the potential of formulating highly biocompatible niosomes in the future of nanomedicine, enabling more precise and effective treatment of cancers.
Collapse
Affiliation(s)
| | - Farzaneh Tafvizi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran.
| | - Vahid Naseh
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Masoud Salehipour
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| |
Collapse
|
5
|
Kang DY, Park S, Song KS, Bae SW, Lee JS, Jang KJ, Park YM. Anticancer Effects of 6-Gingerol through Downregulating Iron Transport and PD-L1 Expression in Non-Small Cell Lung Cancer Cells. Cells 2023; 12:2628. [PMID: 37998363 PMCID: PMC10670414 DOI: 10.3390/cells12222628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Iron homeostasis is considered a key factor in human metabolism, and abrogation in the system could create adverse effects, including cancer. Moreover, 6-gingerol is a widely used bioactive phenolic compound with anticancer activity, and studies on its exact mechanisms on non-small cell lung cancer (NSCLC) cells are still undergoing. This study aimed to find the mechanism of cell death induction by 6-gingerol in NSCLC cells. Western blotting, real-time polymerase chain reaction, and flow cytometry were used for molecular signaling studies, and invasion and tumorsphere formation assay were also used with comet assay for cellular processes. Our results show that 6-gingerol inhibited cancer cell proliferation and induced DNA damage response, cell cycle arrest, and apoptosis in NSCLC cells, and cell death induction was found to be the mitochondrial-dependent intrinsic apoptosis pathway. The role of iron homeostasis in the cell death induction of 6-gingerol was also investigated, and iron metabolism played a vital role in the anticancer ability of 6-gingerol by downregulating EGFR/JAK2/STAT5b signaling or upregulating p53 and downregulating PD-L1 expression. Also, 6-gingerol induced miR-34a and miR-200c expression, which may indicate regulation of PD-L1 expression by 6-gingerol. These results suggest that 6-gingerol could be a candidate drug against NSCLC cells and that 6-gingerol could play a vital role in cancer immunotherapy.
Collapse
Affiliation(s)
- Dong Young Kang
- Department of Immunology, School of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Sanghyeon Park
- Department of Immunology, School of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Kyoung Seob Song
- Department of Medical Science, Kosin University College of Medicine, Busan 49267, Republic of Korea
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea
| | - Jeong-Sang Lee
- Department of Functional Foods and Biotechnology, College of Medical Sciences, Jeonju University, Jeonju 55069, Republic of Korea
| | - Kyoung-Jin Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yeong-Min Park
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
6
|
Chen GQ, Nan Y, Huang SC, Ning N, Du YH, Lu DD, Yang YT, Meng FD, Yuan L. Research progress of ginger in the treatment of gastrointestinal tumors. World J Gastrointest Oncol 2023; 15:1835-1851. [DOI: 10.4251/wjgo.v15.i11.1835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer seriously endangers human health. Gastrointestinal cancer is the most common and major malignant tumor, and its morbidity and mortality are gradually increasing. Although there are effective treatments such as radiotherapy and chemotherapy for gastrointestinal tumors, they are often accompanied by serious side effects. According to the traditional Chinese medicine and food homology theory, many materials are both food and medicine. Moreover, food is just as capable of preventing and treating diseases as medicine. Medicine and food homologous herbs not only have excellent pharmacological effects and activities but also have few side effects. As a typical medicinal herb with both medicinal and edible uses, some components of ginger have been shown to have good efficacy and safety against cancer. A mass of evidence has also shown that ginger has anti-tumor effects on digestive tract cancers (such as gastric cancer, colorectal cancer, liver cancer, laryngeal cancer, and pancreatic cancer) through a variety of pathways. The aim of this study is to investigate the mechanisms of action of the main components of ginger and their potential clinical applications in treating gastrointestinal tumors.
Collapse
Affiliation(s)
- Guo-Qing Chen
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- School of Clinical Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ting Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Fan-Di Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
7
|
Wu QH, Luo L, Luo Q, Hong T, Xu L, Ma Q, Liu L, Liu ZY. Dietary ginger polysaccharides (Gps) improve symptoms in hyperlipidemia rats via alterations in gut microbiota. Heliyon 2023; 9:e17534. [PMID: 37456047 PMCID: PMC10345252 DOI: 10.1016/j.heliyon.2023.e17534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The aim of this research is to investigate lipid-lowering influence of dietary ginger (Zingier officinales Rocs) polysaccharides (GPS) on hyperlipidemia rats. Rat models with hyperlipidemia was established by high-fat food diet (HFD). Comparing to GP-negative model group, GPS attenuated several effects of HFD feeding, including the levels of blood lipid biochemistry, serum inflammatory markers (tumor necrosis factor TNF-a, interleukin IL-6), antioxidant capacity (superoxide dismutase SOD, glutathione peroxidase GSH-Px, total antioxidant capacity T-AOC, propylene dialdehyde MDA), uric acid and immune index. 16 S rDNA gene sequencing of fecal samples showed that GPS increased the growth of Akkermansia muciniphila and decreased the proportion of Firmicutes to Bacteroidetes; This changes in microbial community structure can help prevent diet-induced metabolic disease. These results suggest that GPs may act on the gut, changing the structure of the gut microbial community, thereby reducing intestinal and systemic inflammation, thus improved metabolic outcomes.
Collapse
Affiliation(s)
- Qing-hua Wu
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Liming Luo
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Qi Luo
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Tao Hong
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Lei Xu
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Qiuting Ma
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| | - Li Liu
- Nanchang Medical University, Nanchang 330006, China
| | - Zhi-yong Liu
- Jiangxi University of Traditional Chinese Medicine2019, Nanchang 330004, China
| |
Collapse
|
8
|
Garza-Cadena C, Ortega-Rivera DM, Machorro-García G, Gonzalez-Zermeño EM, Homma-Dueñas D, Plata-Gryl M, Castro-Muñoz R. A comprehensive review on Ginger (Zingiber officinale) as a potential source of nutraceuticals for food formulations: Towards the polishing of gingerol and other present biomolecules. Food Chem 2023; 413:135629. [PMID: 36753787 DOI: 10.1016/j.foodchem.2023.135629] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Currently, ginger is one the most consumed plants when dealing with the treatments of various illnesses. So far, it is known that various biologically active molecules, such as gingerols, shogaols and zingerone, among others, are the main responsible for specific biological activities, opening a new window for its utilization as a nutraceutical in foods. In pioneering extraction processes, solvent extraction has been initially used for these applications; however, the drawbacks of this typical extraction method compared with other emergent separation techniques make it possible for the exploration of new extraction pathways, including microwave, ultrasound, supercritical, subcritical and pressurized-assisted extraction, along with three phase partitioning, high-speed counter current chromatography and magnetic solid phase extraction. To the best of our knowledge, there is no report documenting the recent studies and cases of study in this field. Therefore, we comprehensively review the progress and the latest findings (over the last five years) on research developments, including patents and emerging extraction methods, aiming at the purification of biologically active molecules (gingerols, shogaols and zingerone) contained in ginger. Over the course of this review, particular emphasis is devoted to breakthrough strategies and meaningful outcomes in ginger components extraction. Finally, dosage and safety concerns related to ginger extracts are also documented.
Collapse
Affiliation(s)
- Clarissa Garza-Cadena
- Tecnologico de Monterrey, Campus Monterrey, Av. Eugenio Garza Sada, Sur 2501 Sur, Tecnológico, 64849 Monterrey, NL, Mexico
| | - Daniela Marian Ortega-Rivera
- Tecnologico de Monterrey, Campus Ciudad de México, Prol. Canal de Miramontes, Coapa, San Bartolo el Chico, Tlalpan, 14380 Ciudad de México, CDMX, Mexico
| | - Gerson Machorro-García
- Tecnologico de Monterrey, Campus Monterrey, Av. Eugenio Garza Sada, Sur 2501 Sur, Tecnológico, 64849 Monterrey, NL, Mexico
| | - Eloy Mauricio Gonzalez-Zermeño
- Tecnologico de Monterrey, Campus Ciudad de México, Prol. Canal de Miramontes, Coapa, San Bartolo el Chico, Tlalpan, 14380 Ciudad de México, CDMX, Mexico
| | - Diego Homma-Dueñas
- Tecnologico de Monterrey, Campus Monterrey, Av. Eugenio Garza Sada, Sur 2501 Sur, Tecnológico, 64849 Monterrey, NL, Mexico
| | - Maksymilian Plata-Gryl
- Gdansk University of Technology, Faculty of Chemistry, Department of Process Engineering and Chemical Technology, G.Narutowicza St. 11/12, 80-233 Gdansk, Poland; Gdansk University of Technology, Faculty of Civil and Environmental Engineering, Department of Sanitary Engineering, 11/12 Narutowicza St, 80-233 Gdansk, Poland
| | - Roberto Castro-Muñoz
- Gdansk University of Technology, Faculty of Civil and Environmental Engineering, Department of Sanitary Engineering, 11/12 Narutowicza St, 80-233 Gdansk, Poland; Tecnologico de Monterrey, Campus Toluca, Av. Eduardo Monroy, Cárdenas 2000 San Antonio Buenavista, 50110 Toluca de Lerdo, Mexico.
| |
Collapse
|
9
|
Salari Z, Khosravi A, Pourkhandani E, Molaakbari E, Salarkia E, Keyhani A, Sharifi I, Tavakkoli H, Sohbati S, Dabiri S, Ren G, Shafie’ei M. The inhibitory effect of 6-gingerol and cisplatin on ovarian cancer and antitumor activity: In silico, in vitro, and in vivo. Front Oncol 2023; 13:1098429. [PMID: 36937441 PMCID: PMC10020515 DOI: 10.3389/fonc.2023.1098429] [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: 11/14/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Background Epithelial ovarian cancer is very common in women and causes hundreds of deaths per year worldwide. Chemotherapy drugs including cisplatin have adverse effects on patients' health. Complementary treatments and the use of herbal medicines can help improve the performance of medicine. 6-Gingerol is the major pharmacologically active component of ginger. In this study, we compared the effects of 6-gingerol, cisplatin, and their combination in apoptotic and angiogenetic activities in silico, in test tubes, and in in vivo assays against two ovarian cancer cell lines: OVCAR-3 and human umbilical vein endothelial cells (HUVECs). Methods The drug-treated cell lines were evaluated for their cytotoxicity, cell cycle, and apoptotic and angiogenetic gene expression changes. Results The proportion of apoptosis treated by 6-gingerol coupled with cisplatin was significantly high. In the evaluation of the cell cycle, the combination therapy also showed a significant promotion of a higher extent of the S sequence. The expression of p53 level, Caspase-8, Bax, and Apaf1 genes was amplified again with combination therapy. Conversely, in both cell lines, the cumulative drug concentrations reduced the expression of VEGF, FLT1, KDR, and Bcl-2 genes. Similarly, in the control group, combination treatment significantly decreased the expression of VEGF, FLT1, KDR, and Bcl-2 genes in comparison to cisplatin alone. Conclusions The findings of the present study demonstrated that the cisplatin and 6-gingerol combination is more effective in inducing apoptosis and suppressing the angiogenesis of ovarian cancer cells than using each drug alone.
Collapse
Affiliation(s)
- Zohreh Salari
- Obstetrics and Gynecology Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
- *Correspondence: Ahmad Khosravi, ; Elham Pourkhandani,
| | - Elham Pourkhandani
- Obstetrics and Gynecology Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- *Correspondence: Ahmad Khosravi, ; Elham Pourkhandani,
| | - Elaheh Molaakbari
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ehsan Salarkia
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Keyhani
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hadi Tavakkoli
- Department of Clinical Science, School of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Samira Sohbati
- Obstetrics and Gynecology Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Afzalipour School of Medicine and Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Guogang Ren
- School of Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom
| | - Mohammad Shafie’ei
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
Žitek T, Kučuk N, Postružnik V, Leitgeb M, Knez Ž, Primožič M, Marevci MK. Synergistic Effect of Supercritical and Ultrasound-Assisted Ginger ( Zingiber officinale Roscoe) Extracts. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11212872. [PMID: 36365323 PMCID: PMC9654478 DOI: 10.3390/plants11212872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 06/01/2023]
Abstract
Proper processing of natural material is crucial to obtain an extract with high content of biologically active components. Dried, grinded ginger roots were extracted by ultrasonic method and supercritical extraction with CO2. The aim of the study was to determine if a mixture of the two types of extracts attained by different methods and solvents exhibits better bioavailability than each extract itself. Therefore, both extracts were analytically evaluated and then mixed in a ratio of 1:1. The supercritical extract (SCG extract) and the mixed extract (mixG extract) had high antioxidant activity (78% and 73%) and total phenols (827 mg/g ext. and 1455 mg/g ext.), which is also consistent with the levels of gingerol (303 mg/g ext. and 271 g/g ext.) and shogaol (111 mg/g ext. and 100 g/g ext.) in the extracts. In comparison to both pure extracts higher levels of total phenols were found in the extract mixG. This could be the reason for the significant inhibition of melanoma cells and antimicrobial potential (against Staphylococcus aureus, Escherichia coli, and Candida albicans). The combination of the extracts resulted in a significant increase in the inhibition of selected microbial and melanoma cells WM-266-4 compared to the control. Cell viability decreased below 60% when mixG extract was applied. Antimicrobial activity has been confirmed.
Collapse
Affiliation(s)
- Taja Žitek
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| | - Nika Kučuk
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| | - Vesna Postružnik
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| | - Maja Leitgeb
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Željko Knez
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Mateja Primožič
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| | - Maša Knez Marevci
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
11
|
Role of Plant-Derived Active Constituents in Cancer Treatment and Their Mechanisms of Action. Cells 2022; 11:cells11081326. [PMID: 35456005 PMCID: PMC9031068 DOI: 10.3390/cells11081326] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
Abstract
Despite significant technological advancements in conventional therapies, cancer remains one of the main causes of death worldwide. Although substantial progress has been made in the control and treatment of cancer, several limitations still exist, and there is scope for further advancements. Several adverse effects are associated with modern chemotherapy that hinder cancer treatment and lead to other critical disorders. Since ancient times, plant-based medicines have been employed in clinical practice and have yielded good results with few side effects. The modern research system and advanced screening techniques for plants’ bioactive constituents have enabled phytochemical discovery for the prevention and treatment of challenging diseases such as cancer. Phytochemicals such as vincristine, vinblastine, paclitaxel, curcumin, colchicine, and lycopene have shown promising anticancer effects. Discovery of more plant-derived bioactive compounds should be encouraged via the exploitation of advanced and innovative research techniques, to prevent and treat advanced-stage cancers without causing significant adverse effects. This review highlights numerous plant-derived bioactive molecules that have shown potential as anticancer agents and their probable mechanisms of action and provides an overview of in vitro, in vivo and clinical trial studies on anticancer phytochemicals.
Collapse
|
12
|
|
13
|
Keyhani A, Sharifi I, Salarkia E, Khosravi A, Tavakoli Oliaee R, Babaei Z, Ghasemi Nejad Almani P, Hassanzadeh S, Kheirandish R, Mostafavi M, Hakimi Parizi M, Alahdin S, Sharifi F, Dabiri S, Shamsi Meymandi S, Khamesipour A, Jafarzadeh A, Bamorovat M. In vitro and in vivo therapeutic potentials of 6-gingerol in combination with amphotericin B for treatment of Leishmania major infection: Powerful synergistic and multifunctional effects. Int Immunopharmacol 2021; 101:108274. [PMID: 34688150 DOI: 10.1016/j.intimp.2021.108274] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022]
Abstract
The ongoing conventional drugs for leishmaniasis treatment are insufficient. The present study aimed to assess 6-gingerol alone and in combination with amphotericin B on Leishmania major stages using experimental and in vivo murine models. Here, arrays of experimental approaches were designed to monitor and evaluate the 6-gingerol potential therapeutic outcomes. The binding affinity of 6-gingerol and IFN-γ was the basis for docking conformations. 6-Gingerol combined with amphotericin B represented a safe mixture, extremely leishmanicidal, a potent antioxidant, induced a remarkable apoptotic index, significantly increased the expression of the Th1-related cytokines (IL-12p40, IFN-γ, and TNF- α), iNOS, and transcription factors (STAT1, c-Fos, and Elk-1). In contrast, the expression of the Th2-related cytokines was significantly downregulated (p < 0.001). This combination was also potent when the lesion appearance was evaluated following three weeks of treatment. The histopathological and immunohistochemical patterns of the murine model represented clusters of CD4+ and CD8+ T lymphocytes which compressed and deteriorated the macrophages harboring Leishman bodies. The primary mode of action of 6-gingerol and amphotericin B involved broad mechanistic insights providing a coherent basis for further clinical study as a potential drug candidate for CL. In conclusion, 6-gingerol with amphotericin B synergistically exerted anti-leishmanial activity in vitro and in vivo and potentiated macrophages' leishmanicidal activity, modulated Th1- and Th2-related phenotypes improved the histopathological changes in the BALB/c mice infected with L. major. They elevated the leukocyte infiltration into the lesions. Therefore, this combination should be considered for treating volunteer patients with CL in clinical studies.
Collapse
Affiliation(s)
- Alireza Keyhani
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ehsan Salarkia
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Zahra Babaei
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Saeid Hassanzadeh
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Reza Kheirandish
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mashid Mostafavi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Hakimi Parizi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Sodabeh Alahdin
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sharifi
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Department of Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Simin Shamsi Meymandi
- Department of Dermatology, Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Sp N, Kang DY, Jo ES, Lee JM, Bae SW, Jang KJ. Pivotal Role of Iron Homeostasis in the Induction of Mitochondrial Apoptosis by 6-Gingerol Through PTEN Regulated PD-L1 Expression in Embryonic Cancer Cells. Front Oncol 2021; 11:781720. [PMID: 34804985 PMCID: PMC8595921 DOI: 10.3389/fonc.2021.781720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSCs with natural compounds is a promising approach as it suppresses cancer recurrence with fewer adverse effects. 6-Gingerol is an active component of ginger, which exhibits well-known anti-cancer activities. This study determined the mechanistic aspects of cell death induction by 6-gingerol. To analyze cellular processes, we used Western blot and real-time qPCR for molecular signaling studies and conducted flow cytometry. Our results suggested an inhibition of CSC marker expression and Wnt/β-catenin signaling by 6-gingerol in NCCIT and NTERA-2 cells. 6-Gingerol induced reactive oxygen species generation, the DNA damage response, cell cycle arrest, and the intrinsic pathway of apoptosis in embryonic CSCs. Furthermore, 6-gingerol inhibited iron metabolism and induced PTEN, which both played vital roles in the induction of cell death. The activation of PTEN resulted in the inhibition of PD-L1 expression through PI3K/AKT/p53 signaling. The induction of PTEN also mediated the downregulation of microRNAs miR-20b, miR-21, and miR-130b to result in PD-L1 suppression by 6-gingerol. Hence, 6-gingerol may be a promising candidate to target CSCs by regulating PTEN-mediated PD-L1 expression.
Collapse
Affiliation(s)
- Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| | - Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| | - Eun Seong Jo
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si, South Korea
| | - Jin-Moo Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si, South Korea.,SK Bioscience, Seongnam-si, South Korea
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju, South Korea
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| |
Collapse
|
15
|
Ahmed AS, Soliman MG. Protective Role of 4-(4-Hydroxy-3-methoxyphenyl)-2-Butanone on Prostatic Cells Hyperplasia of Rats and Human, 5α-reductase Inhibition Pathway. J Microsc Ultrastruct 2021; 9:164-169. [PMID: 35070691 PMCID: PMC8751679 DOI: 10.4103/jmau.jmau_37_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 05/17/2020] [Accepted: 09/03/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Prostate gland is an exocrine gland that could be affected by various pathological conditions. Benign prostatic hyperplasia (BPH) is an age-dependent medical condition caused by increased activity of 5α-reductase enzyme (5α-R). Medical treatment by finasteride is considered during treatment, but it has unavoidable side effects. Hence, there is an increasing need to use natural ingredients for BPH treatment. Gingerol oil (ginger extract) is transferred by heating into zingerone. Recent studies reported the effect of zingerone on prostate cancer cells. AIMS AND OBJECTIVES The aim of the present research is to investigate the protective effect of zingerone against BPH. MATERIALS AND METHODS Sixty male Albino Wistar rats were divided into three groups: control group, prostatic hyperplasia group treated with saline, and prostatic hyperplasia group treated with zingerone (PH-Z-G). At day 28, all rats were sacrificed, epididymis and prostate samples were collected for histopathological examination and Western blotting for androgen receptors (ARs) proteins and steroid 5 alpha-reductase 1 (SRD5A1). Human RWPE-1 prostatic cell line was assessed for viability and cycle after treated with zingerone 500 μg/day for 10 days. RESULTS PH-S group showed significant (P < 0.05) thickening of connective tissue septa associated with narrowing of acinar lumen. PH-Z group showed regain of the normal histological feature. SRD5A1 and AR expression was significantly (P < 0.05) reduced in PH-Z group in comparison with PH-S group. Cell line proliferation was significantly reduced after application of zingerone with G2/M cell cycle arrest. CONCLUSION Our results showed that natural herbal zingerone decreased the prostatic tissue levels of (5α reductase and AR) in rat BPH model, which could be a promising herbal medicine for BPH treatment. Further human clinical trials are required.
Collapse
Affiliation(s)
- Ahmed S. Ahmed
- Department of Anatomy and Embryology, College of Medicine, Tanta University, Tanta, Egypt
| | | |
Collapse
|
16
|
Wishart G, Gupta P, Nisbet A, Velliou E, Schettino G. Novel Anticancer and Treatment Sensitizing Compounds against Pancreatic Cancer. Cancers (Basel) 2021; 13:2940. [PMID: 34208295 PMCID: PMC8231164 DOI: 10.3390/cancers13122940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
The isolation of chemical compounds from natural origins for medical application has played an important role in modern medicine with a range of novel treatments having emerged from various natural forms over the past decades. Natural compounds have been exploited for their antioxidant, antimicrobial and antitumor capabilities. Specifically, 60% of today's anticancer drugs originate from natural sources. Moreover, the combination of synthetic and natural treatments has shown applications for (i) reduced side effects, (ii) treatment sensitization and (iii) reduction in treatment resistance. This review aims to collate novel and natural compounds that are being explored for their preclinical anticancer, chemosensitizing and radiosensitizing effects on Pancreatic Ductal Adenocarcinoma (PDAC), which is a lethal disease with current treatments being inefficient and causing serve side effects. Two key points are highlighted by this work: (i) the availability of a range of natural compounds for potentially new therapeutic approaches for PDAC, (ii) potential synergetic impact of natural compounds with advanced chemo- and radio-therapeutic modalities for PDAC.
Collapse
Affiliation(s)
- Gabrielle Wishart
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, UK;
| | - Eirini Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Centre for 3D Models of Health and Disease, UCL-Division of Surgery and Interventional Science, Charles Bell House, 43-45 Foley Street, Fitzrovia, London W1W 7TY, UK
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
- National Physical Laboratory, Teddington TW11 0LW, UK
| |
Collapse
|
17
|
Sp N, Kang DY, Lee JM, Bae SW, Jang KJ. Potential Antitumor Effects of 6-Gingerol in p53-Dependent Mitochondrial Apoptosis and Inhibition of Tumor Sphere Formation in Breast Cancer Cells. Int J Mol Sci 2021; 22:4660. [PMID: 33925065 PMCID: PMC8124719 DOI: 10.3390/ijms22094660] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Hormone-specific anticancer drugs for breast cancer treatment can cause serious side effects. Thus, treatment with natural compounds has been considered a better approach as this minimizes side effects and has multiple targets. 6-Gingerol is an active polyphenol in ginger with various modalities, including anticancer activity, although its mechanism of action remains unknown. Increases in the level of reactive oxygen species (ROS) can lead to DNA damage and the induction of DNA damage response (DDR) mechanism, leading to cell cycle arrest apoptosis and tumorsphere suppression. Epidermal growth factor receptor (EGFR) promotes tumor growth by stimulating signaling of downstream targets that in turn activates tumor protein 53 (p53) to promote apoptosis. Here we assessed the effect of 6-gingerol treatment on MDA-MB-231 and MCF-7 breast cancer cell lines. 6-Gingerol induced cellular and mitochondrial ROS that elevated DDR through ataxia-telangiectasia mutated and p53 activation. 6-Gingerol also induced G0/G1 cell cycle arrest and mitochondrial apoptosis by mediating the BAX/BCL-2 ratio and release of cytochrome c. It also exhibited a suppression ability of tumorsphere formation in breast cancer cells. EGFR/Src/STAT3 signaling was also determined to be responsible for p53 activation and that 6-gingerol induced p53-dependent intrinsic apoptosis in breast cancer cells. Therefore, 6-gingerol may be used as a candidate drug against hormone-dependent breast cancer cells.
Collapse
Affiliation(s)
- Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
| | - Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
| | - Jin-Moo Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju 28159, Korea;
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Korea;
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
| |
Collapse
|
18
|
Zivarpour P, Nikkhah E, Maleki Dana P, Asemi Z, Hallajzadeh J. Molecular and biological functions of gingerol as a natural effective therapeutic drug for cervical cancer. J Ovarian Res 2021; 14:43. [PMID: 33706784 PMCID: PMC7953815 DOI: 10.1186/s13048-021-00789-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer is one of the most common and important gynecological cancers, which has a global concern with an increasing number of patients and mortality rates. Today, most women in the world who suffer from cervical cancer are developing advanced stages of the disease. Smoking and even exposure to secondhand smoke, infections caused by the human papillomavirus, immune system dysfunction and high-risk individual-social behaviors are among the most important predisposing factors for this type of cancer. In addition, papilloma virus infection plays a more prominent role in cervical cancer. Surgery, chemotherapy or radical hysterectomy, and radiotherapy are effective treatments for this condition, the side effects of these methods endanger a person's quality of life and cause other problems in other parts of the body. Studies show that herbal medicines, including taxol, camptothecin and combretastatins, have been shown to be effective in treating cervical cancer. Ginger (Zingiber officinale, Zingiberaceae) is one of the plants with valuable compounds such as gingerols, paradols and shogoals, which is a rich source of antioxidants, anti-cancer and anti-inflammatory agents. Numerous studies have reported the therapeutic effects of this plant through various pathways in cervical cancer. In this article, we look at the signaling mechanisms and pathways in which ginger is used to treat cervical cancer.
Collapse
Affiliation(s)
- Parinaz Zivarpour
- Department of Biological sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Elhameh Nikkhah
- Medicinal Plants Research Cent Maragheh University of Medical Sciences, Maragheh, Iran
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
19
|
Xu S, Zhang H, Liu T, Wang Z, Yang W, Hou T, Wang X, He D, Zheng P. 6-Gingerol suppresses tumor cell metastasis by increasing YAP ser127 phosphorylation in renal cell carcinoma. J Biochem Mol Toxicol 2021; 35:e22609. [PMID: 32926756 DOI: 10.1002/jbt.22609] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/15/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
According to the World Health Organization, the incidence and mortality rates of renal cell carcinoma (RCC) are rapidly increasing worldwide. Serious side effects caused by immune therapy and resistance to targeted drug therapy are urgent clinical problems facing kidney treatment. There is increasing global interest in developing natural products with a reduced number of side effects as adjunctive therapeutic options for RCC. Ginger is a spice and herbal remedy used worldwide, and 6-gingerol is a major pharmacologically active ingredient in ginger. In our study, we found that 6-gingerol suppressed RCC cell migration and metastasis in vitro and in vivo. Moreover, reduction in MMP2, Slug, and Vimentin protein levels was observed following 6-gingerol treatment of 786-O and ACHN cells. Furthermore, we revealed the mechanisms underlying the ability of 6-gingerol to inhibit RCC cell migration and metastasis. 6-Gingerol increased yes-associated protein (YAP)ser127 phosphorylation and reduced YAP levels in cell nuclei. We also used a series of loss-of-function and gain-of-function experiments to support our results. Western blot results showed that MMP2, Slug, and Vimentin protein expression was downregulated in YAP-silenced cells and upregulated in YAP-overexpressing cells. Transwell data demonstrated that YAP suppressed RCC migration ability. Immunofluorescence images showed that 6-gingerol decreased YAP levels, leading to disordered F-actin and a reduction in cell lamellipodia. Overall, our results indicated that 6-gingerol is a potential antimetastatic compound for use in kidney therapy.
Collapse
Affiliation(s)
- Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Haibao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Zixi Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Wenjie Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Tao Hou
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- The First Affiliated Hospital of Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Pengsheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
20
|
Sun Y, Ren J, Wang F. [6]-Gingerol impedes 7,12-dimethylbenz(a)anthracene-induced inflammation and cell proliferation-associated hamster buccal pouch carcinogenesis through modulating Nrf2 signaling events. J Biochem Mol Toxicol 2020; 35:e22689. [PMID: 33347680 DOI: 10.1002/jbt.22689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/22/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
The present study examines the chemopreventive role of [6]-gingerol, an active component of ginger, on 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis models. The HBP has been developed with an addition of 0.5% of DMBA to the HBP area three times per week, up to the end of the 16th experimental week. At the end of the experiment, we noticed 100% tumor incidence and precancerous lesions, such as dysplasia, hyperplasia, keratosis, and well-differentiated squamous cell carcinoma, in DMBA-induced HBP. Furthermore, we observed that [6]-gingerol inhibited the increased thiobarbituric acid-reactive substances and decreased antioxidant levels in DMBA-induced hamsters. Moreover, [6]-gingerol inhibits DMBA-exposed over expression of inflammatory markers (inducible nitric oxide synthase, interleukin [IL]-1β, IL-6, cyclooxygenase-2, and tumor necrosis factor-α) and cell proliferation markers (cyclin D1, proliferating cell nuclear antigen); induces proapoptotic markers in HBP. Nuclear factor erythroid-2-related factor-2 (Nrf2) is a major antioxidant transcription factor, which regulates the antioxidant gene-dependent scavenge of tumor proliferation and apoptosis. Overexpression of Nrf2 signaling plays a pivotal role and can be a novel target in preventing carcinogenesis. In this study, [6]-gingerol restores the DMBA-induced depletion of Nrf2 signaling and thereby prevents buccal pouch carcinogenesis in hamsters. These results point out that [6]-gingerol impedes the responses of inflammatory and cell proliferation-associated progression of cancer through the action of Nrf2 signaling.
Collapse
Affiliation(s)
- Yugang Sun
- Oral and maxillofacial surgery, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Jinmin Ren
- Health Management Center, Binzhou Municipal Hospital of Traditional Chinese Medicine, Binzhou, Shandong, China
| | - Fang Wang
- Department of Oncology, The Second People Hospital of Dezhou, Dezhou, Shandong, China
| |
Collapse
|
21
|
Alamri HS, Alsughayyir J, Akiel M, Al-Sheikh YA, Basudan AM, Dera A, Barhoumi T, Basuwdan AM, Alfhili MA. Stimulation of calcium influx and CK1α by NF-κB antagonist [6]-Gingerol reprograms red blood cell longevity. J Food Biochem 2020; 45:e13545. [PMID: 33145778 DOI: 10.1111/jfbc.13545] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022]
Abstract
Chemotherapy-induced anemia (CIA) is a major obstacle in cancer management. Although the mechanisms governing CIA are poorly understood, recent efforts have identified suicidal erythrocyte (red blood cell, RBC) death as a possible cause of CIA. [6]-Gingerol (GNG), a polyphenol extracted from Zingiber officinale plant, exhibits a wide array of biological activities including antimicrobial, antioxidant, anti-inflammatory, immunomodulatory, and anticancer activities, in vitro and in vivo. However, the potential toxicity of GNG to human RBCs remains unexplored. RBCs from heparinized blood were isolated by centrifugation and exposed to antitumor concentrations (10-100 µM) of GNG for 24 hr at 37°C. Hemolysis was calculated from hemoglobin leakage in the supernatant (λmax = 405 nm), while cytofluorometric analysis of eryptosis employed Annexin-V-FITC to detect phosphatidylserine (PS) exposure, forward scatter (FSC) to estimate cell volume, Fluo4/AM to measure calcium activity, and H2 DCFDA to assess oxidative stress. Moreover, zVAD(OMe)-FMK, SB203580, necrostatin-2, staurosporin, and D4476 were used to identify signaling pathways responsive to GNG. GNG induced significant hemolysis at 100 µM, independently of extracellular calcium, and increased Annexin-V-FITC fluorescence that was thoroughly abrogated without extracellular calcium. GNG also enhanced Fluo4 fluorescence and reduced FSC, but had no significant effect on DCF fluorescence. Importantly, the presence of D4476 significantly attenuated GNG-induced hemolysis. In conclusion, GNG stimulates premature RBC death characterized by loss of membrane asymmetry, elevated cytosolic calcium, cell shrinkage, and casein kinase 1α activation. Blocking the activity of calcium channels or CK1α may, therefore, ameliorate the toxic effects of GNG on RBCs. PRACTICAL APPLICATIONS: This report presents a safety assessment of GNG as a chemotherapeutic agent and highlights the novel toxicity of GNG to human RBCs. Our findings provide novel insights that may lead to more efficient utilization of GNG in chemotherapy. Specifically, our data revealed the involvement of calcium channels and casein kinase 1α in mediating GNG-induced premature RBC death, and, therefore, inverse agonists or inhibitors of either pathway may be used as pharmaceutical adjuvants to attenuate the toxic effects of GNG.
Collapse
Affiliation(s)
- Hassan S Alamri
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia.,King Abdullah International Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Maaged Akiel
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia.,King Abdullah International Research Center (KAIMRC), Riyadh, Saudi Arabia.,Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yazeed A Al-Sheikh
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed M Basudan
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ayed Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.,Research Centre of Advanced Materials, King Khalid University, Abha, Saudi Arabia
| | - Tlili Barhoumi
- Medical Core Facility and Research Platforms, King Abdullah International Research Center (KAIMRC), King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Abdulrahman M Basuwdan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Mohammad A Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Puckett DL, Alquraishi M, Alani D, Chahed S, Donohoe D, Voy B, Whelan J, Bettaieb A. Zyflamend induces apoptosis in pancreatic cancer cells via modulation of the JNK pathway. Cell Commun Signal 2020; 18:126. [PMID: 32795297 PMCID: PMC7427957 DOI: 10.1186/s12964-020-00609-7] [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: 12/18/2019] [Accepted: 06/08/2020] [Indexed: 12/16/2022] Open
Abstract
Background Current pharmacological therapies and treatments targeting pancreatic neuroendocrine tumors (PNETs) have proven ineffective, far too often. Therefore, there is an urgent need for alternative therapeutic approaches. Zyflamend, a combination of anti-inflammatory herbal extracts, that has proven to be effective in various in vitro and in vivo cancer platforms, shows promise. However, its effects on pancreatic cancer, in particular, remain largely unexplored. Methods In the current study, we investigated the effects of Zyflamend on the survival of beta-TC-6 pancreatic insulinoma cells (β-TC6) and conducted a detailed analysis of the underlying molecular mechanisms. Results Herein, we demonstrate that Zyflamend treatment decreased cell proliferation in a dose-dependent manner, concomitant with increased apoptotic cell death and cell cycle arrest at the G2/M phase. At the molecular level, treatment with Zyflamend led to the induction of ER stress, autophagy, and the activation of c-Jun N-terminal kinase (JNK) pathway. Notably, pharmacological inhibition of JNK abrogated the pro-apoptotic effects of Zyflamend. Furthermore, Zyflamend exacerbated the effects of streptozotocin and adriamycin-induced ER stress, autophagy, and apoptosis. Conclusion The current study identifies Zyflamend as a potential novel adjuvant in the treatment of pancreatic cancer via modulation of the JNK pathway. Video abstract
Collapse
Affiliation(s)
- Dexter L Puckett
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA
| | - Dina Alani
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA
| | - Samah Chahed
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA
| | - Dallas Donohoe
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA
| | - Brynn Voy
- Tennessee Agricultural Experiment Station, University of Tennessee Institute of Agriculture, Knoxville, TN, 37996-0840, USA.,Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996-0840, USA
| | - Jay Whelan
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA.,Tennessee Agricultural Experiment Station, University of Tennessee Institute of Agriculture, Knoxville, TN, 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, 1215 Cumberland Avenue, 229 Jessie Harris Building, Knoxville, TN, 37996-0840, USA. .,Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996-0840, USA. .,Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996-0840, USA.
| |
Collapse
|
23
|
Yapasert R, Sripanidkulchai B, Teerachaisakul M, Banchuen K, Banjerdpongchai R. Anticancer effects of a traditional Thai herbal recipe Benja Amarit extracts against human hepatocellular carcinoma and colon cancer cell by targeting apoptosis pathways. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112732. [PMID: 32142865 DOI: 10.1016/j.jep.2020.112732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE A preparation of Benja Amarit (BJA) has been effectively used in folk medicine to treat diseases related to the liver and colon and forms of cancer for hundreds of years in Thailand. However, there has not been any research on BJA with regard to its anticancer activity against human hepatocellular carcinoma and colon cancer cells. AIM OF THE STUDY This study was to obtain the scientific supports for the traditional usage in anticancer potential of BJA extracts on hepatocellular carcinoma and colon cancer. MATERIALS AND METHODS The cytotoxic effects were determined using MTT assay. Apoptosis was quantitated by annexin V-FITC/PI staining. Caspases activities were measured by using specific substrates and colorimetric analysis. The protein expressions were determined by Western blot analysis. Reactive oxygen species (ROS) generation, mitochondrial transmembrane potential, and calcium ion levels were measured by specific fluorescence probes and flow cytometry. The chick embryo chorioallantoic membrane model has been used to study the in vivo anticancer activity. The phytochemical identification was performed by GC-MS and LC-MS. RESULTS Notably, 95% (BJA-95) and 50% (BJA-50) ethanolic extract of BJA inhibited hepatocellular carcinoma and colon cancer cell viability in a dose-dependent manner. While, the water extract of BJA (BJA-W) was not found to be toxic to both kinds of cancer cell lines. BJA extract induced both the extrinsic and intrinsic or mitochondria-mediated apoptosis pathways. Moreover, BJA-95 caused ROS generation and endoplasmic reticulum stress-mediated apoptosis. The extract exhibited the growth inhibitory effects on cancer cells in vivo. Phytochemical analysis revealed that the major active compounds were piperine, xanthotoxol and dihydrogambogic acid. CONCLUSION This study is the first to demonstrate anticancer efficiency of BJA extracts on human cancer cells. We consider BJA extract to be a potentially alternative cancer treatment and to be a promising candidate in the future development of antitumor agents.
Collapse
Affiliation(s)
- Rittibet Yapasert
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Bungorn Sripanidkulchai
- Center for Research and Development of Herbal Health Products, Thailand; Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Monthaka Teerachaisakul
- Thai Traditional Medicine Research Institute, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Bangkok, 10100, Thailand
| | - Kamonwan Banchuen
- Thai Traditional Medicine Research Institute, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Bangkok, 10100, Thailand
| | - Ratana Banjerdpongchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
24
|
Ahmad R, Khan MA, Srivastava A, Gupta A, Srivastava A, Jafri TR, Siddiqui Z, Chaubey S, Khan T, Srivastava AK. Anticancer Potential of Dietary Natural Products: A Comprehensive Review. Anticancer Agents Med Chem 2020; 20:122-236. [DOI: 10.2174/1871520619666191015103712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Nature is a rich source of natural drug-like compounds with minimal side effects. Phytochemicals
better known as “Natural Products” are found abundantly in a number of plants. Since time immemorial, spices
have been widely used in Indian cuisine as flavoring and coloring agents. Most of these spices and condiments
are derived from various biodiversity hotspots in India (which contribute 75% of global spice production) and
form the crux of India’s multidiverse and multicultural cuisine. Apart from their aroma, flavor and taste, these
spices and condiments are known to possess several medicinal properties also. Most of these spices are mentioned
in the Ayurveda, the indigenous system of medicine. The antimicrobial, antioxidant, antiproliferative,
antihypertensive and antidiabetic properties of several of these natural products are well documented in
Ayurveda. These phytoconstituemts are known to act as functional immunoboosters, immunomodulators as well
as anti-inflammatory agents. As anticancer agents, their mechanistic action involves cancer cell death via induction
of apoptosis, necrosis and autophagy. The present review provides a comprehensive and collective update
on the potential of 66 commonly used spices as well as their bioactive constituents as anticancer agents. The
review also provides an in-depth update of all major in vitro, in vivo, clinical and pharmacological studies done
on these spices with special emphasis on the potential of these spices and their bioactive constituents as potential
functional foods for prevention, treatment and management of cancer.
Collapse
Affiliation(s)
- Rumana Ahmad
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Mohsin A. Khan
- Chancellor, Era University, Sarfarazganj, Hardoi Road, Lucknow-226003, UP, India
| | - A.N. Srivastava
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Anamika Gupta
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Aditi Srivastava
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tanvir R. Jafri
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Zainab Siddiqui
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Sunaina Chaubey
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow 226026, UP, India
| | - Arvind K. Srivastava
- Department of Food and Nutrition, Era University, Sarfarazganj, Lucknow-226003, UP, India
| |
Collapse
|
25
|
de Lima Silva WC, Conti R, de Almeida LC, Morais PAB, Borges KB, Júnior VL, Costa-Lotufo LV, de Souza Borges W. Novel [6]-gingerol Triazole Derivatives and their Antiproliferative Potential against Tumor Cells. Curr Top Med Chem 2020; 20:161-169. [PMID: 31880263 DOI: 10.2174/1568026620666191227125507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 11/25/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Effective cancer treatment is a major public health challenge. The limitations of current therapies and their adverse effects reduce the efficacy of treatment, leading to significant mortality rates worldwide. Moreover, natural product chemistry occupies a prominent role in the search for new treatment alternatives, by contributing a spectrum of chemical structures that may potentially yield new bioactive compounds. The compound [6]-gingerol (1) is the main active substance in ginger (Zingiber officinale) and several studies have shown it to produce beneficial effects, including antitumor activity. OBJECTIVE This work aims to obtain new gingerol derivatives with cytotoxic activity. METHODS [6]-gingerol was isolated and its derivatives were produced using click chemistry, obtaining eight new compounds. All chemical structures were determined by means of IR, NMR and HRMS data, and cytotoxicity was evaluated in the HCT 116 (colon carcinoma) and MCF-7 (breast carcinoma) cell lines at concentrations of 5 µmol L-1 and 50 µmol L-1. RESULTS At 50 µmol L-1, more than 70% inhibition of cell growth was achieved with compounds 2e, 2g against HCT 116, and 2b, 2d, 2e, 2f and 2g against MCF-7. CONCLUSION The obtained compounds showed only moderate cytotoxic activity. However, the products with substituents occupying the meta position in relation to the triazole ring showed increased cytotoxic properties. The brominated compound (2g) showed the strongest activity, inhibiting cell proliferation by 87%.
Collapse
Affiliation(s)
- William Cezar de Lima Silva
- Programa de Pos-Graduacao em Quimica, Departamento de Quimica, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Raphael Conti
- Programa de Pos-Graduacao em Quimica, Departamento de Quimica, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | | | | | - Keyller Bastos Borges
- Departamento de Ciencias Naturais, Universidade Federal de Sao Joao Del Rei, Sao Joao Del Rei, Brazil
| | - Valdemar Lacerda Júnior
- Programa de Pos-Graduacao em Quimica, Departamento de Quimica, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | | | - Warley de Souza Borges
- Programa de Pos-Graduacao em Quimica, Departamento de Quimica, Universidade Federal do Espirito Santo, Vitoria, Brazil
| |
Collapse
|
26
|
Manna I, Das D, Mondal S, Bandyopadhyay M. Potential Pharmacotherapeutic Phytochemicals from Zingiberaceae for Cancer Prevention. PHARMACOTHERAPEUTIC BOTANICALS FOR CANCER CHEMOPREVENTION 2020:221-281. [DOI: 10.1007/978-981-15-5999-0_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
27
|
6-Gingerol induces cell-cycle G1-phase arrest through AKT-GSK 3β-cyclin D1 pathway in renal-cell carcinoma. Cancer Chemother Pharmacol 2019; 85:379-390. [PMID: 31832810 PMCID: PMC7015962 DOI: 10.1007/s00280-019-03999-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/22/2019] [Indexed: 01/10/2023]
Abstract
Purpose 6-Gingerol, a major biochemical and pharmacological active ingredient of ginger, has shown anti-inflammatory and antitumor activities against various cancers. Searching for natural products with fewer side effects for developing adjunctive therapeutic options is necessary. Methods The effects of 6-gingerol on proliferation, colony formation, and cell cycle in RCC cells were detected by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and propidium iodide (PI) staining, respectively. Western blotting, an immunofluorescence assay, and immunohistochemical staining were performed to assess the expression of relevant proteins. A subcutaneous tumor model was set up to investigate the 6-gingerol effects on tumor growth in vivo, and the pharmacokinetics of 6-gingerol in mice were detected by LC/MS assays. Results 6-Gingerol treatment exerted time- and dose-dependent inhibition of the growth and colony formation of ACHN, 786-O, and 769-P cells, leading to a concomitant induction of cell-cycle G1-phase arrest and decrease in Ki-67 expression in the cell nucleus. Western-blotting results showed that 6-gingerol reduces phosphorylation of protein kinase B (AKT) Ser 473, cyclin-dependent kinases (CDK4), and cyclin D1 and, meanwhile, increases glycogen synthase kinase (GSK 3β) protein amount. Furthermore, the efficacy of 6-gingerol was demonstrated in an in vivo murine model of 786-O. Conclusion The above results indicate that 6-gingerol can induce cell-cycle arrest and cell-growth inhibition through the AKT–GSK 3β–cyclin D1 signaling pathway in vitro and in vivo, suggesting that 6-gingerol should be useful for renal-cell carcinoma treatment. Electronic supplementary material The online version of this article (10.1007/s00280-019-03999-9) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
In Search of Panacea-Review of Recent Studies Concerning Nature-Derived Anticancer Agents. Nutrients 2019; 11:nu11061426. [PMID: 31242602 PMCID: PMC6627480 DOI: 10.3390/nu11061426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
Cancers are one of the leading causes of deaths affecting millions of people around the world, therefore they are currently a major public health problem. The treatment of cancer is based on surgical resection, radiotherapy, chemotherapy or immunotherapy, much of which is often insufficient and cause serious, burdensome and undesirable side effects. For many years, assorted secondary metabolites derived from plants have been used as antitumor agents. Recently, researchers have discovered a large number of new natural substances which can effectively interfere with cancer cells’ metabolism. The most famous groups of these compounds are topoisomerase and mitotic inhibitors. The aim of the latest research is to characterize natural compounds found in many common foods, especially by means of their abilities to regulate cell cycle, growth and differentiation, as well as epigenetic modulation. In this paper, we focus on a review of recent discoveries regarding nature-derived anticancer agents.
Collapse
|
29
|
Suppression of colorectal cancer cell growth by combined treatment of 6-gingerol and γ-tocotrienol via alteration of multiple signalling pathways. J Nat Med 2019; 73:745-760. [DOI: 10.1007/s11418-019-01323-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/20/2019] [Indexed: 12/26/2022]
|
30
|
Almatroudi A, Alsahli MA, Alrumaihi F, Allemailem KS, Rahmani AH. Ginger: A Novel Strategy to Battle Cancer through Modulating Cell Signalling Pathways: A Review. Curr Pharm Biotechnol 2019; 20:5-16. [PMID: 30659535 DOI: 10.2174/1389201020666190119142331] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/24/2022]
Abstract
Numerous studies have been performed in understanding the development of cancer. Though, the mechanism of action of genes in the development of cancer remains to be explained. The current mode of treatment of cancer shows adverse effects on normal cells and also alter the cell signalling pathways. However, ginger and its active compound have fascinated research based on animal model and laboratories during the past decade due to its potentiality in killing cancer cells. Ginger is a mixture of various compounds including gingerol, paradol, zingiberene and shogaol and such compounds are the main players in diseases management. Most of the health-promoting effects of ginger and its active compound can be attributed due to its antioxidant and anti-tumour activity. Besides, the active compound of ginger has proven its role in cancer management through its modulatory effect on tumour suppressor genes, cell cycle, apoptosis, transcription factors, angiogenesis and growth factor. In this review, the role of ginger and its active compound in the inhibition of cancer growth through modulating cell signalling pathways will be reviewed and discussed.
Collapse
Affiliation(s)
- 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
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Khaled S Allemailem
- 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
|
31
|
Mansingh DP, O J S, Sali VK, Vasanthi HR. [6]-Gingerol-induced cell cycle arrest, reactive oxygen species generation, and disruption of mitochondrial membrane potential are associated with apoptosis in human gastric cancer (AGS) cells. J Biochem Mol Toxicol 2018; 32:e22206. [PMID: 30091159 DOI: 10.1002/jbt.22206] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 01/29/2023]
Abstract
Ginger (Zingiber officinale Roscoe), a monocotyledonous herb, is widely used as an herbal medicine owing to the phytoconstituents it possesses. In the current study, the quantity of [6]-gingerol, the major phenolic ketone, in the fresh ginger and dried ginger rhizome was found to be 6.11 µg/mg and 0.407 µg/mg. Furthermore, [6]-gingerol was assessed for its antiapoptotic effects in human gastric adenocarcinoma (AGS) cells evidenced by acridine orange/ethidium bromide staining technique and Annexin-V assay. An increase in reactive oxygen species (ROS) generation led to a decrease in mitochondrial membrane potential (MMP) and subsequent induction of apoptosis. Results disclose that perturbations in MMP are associated with deregulation of Bax/Bcl-2 ratio at protein level, which leads to upregulation of cytochrome-c triggering the caspase cascade. These enduringly suggest that [6]-gingerol can be effectively used for targeting the mitochondrial energy metabolism to manage gastric cancer cells.
Collapse
Affiliation(s)
- Debjani P Mansingh
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Sunanda O J
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Veeresh Kumar Sali
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Hannah R Vasanthi
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry, India
| |
Collapse
|
32
|
Haris P, Mary V, Sudarsanakumar C. Probing the interaction of the phytochemical 6-gingerol from the spice ginger with DNA. Int J Biol Macromol 2018; 113:124-131. [PMID: 29454952 DOI: 10.1016/j.ijbiomac.2018.02.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/11/2022]
Abstract
6-Gingerol [5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) decan-3-one], the bio-active ingredient of the popular Indian spice ginger (Zingiber officinale Roscoe), is well-known for its pharmacological and physiological actions. The potent antioxidant, antiemetic, antiulcer, antimicrobial, analgesic, hypoglycemic, antihypertensive, antihyperlipidemic, immunostimulant, anti-inflammatory, cardiotonic, and cancer prevention activities of 6-Gingerol has been investigated and explored. 6-Gingerol is a good candidate for the treatment of various cancers including prostrate, pancreatic, breast, skin, gastrointestinal, pulmonary, and renal cancer. In this study we report for the first time the molecular recognition of 6-Gingerol with calf thymus DNA (ctDNA) through experimental and molecular modeling techniques confirming a minor groove binding mode of 6-Gingerol with ctDNA. Fluorescence and UV-vis spectroscopic studies confirm the complex formation of 6-gingerol with ctDNA. The energetics and thermodynamics of the interaction of 6-Gingerol with ctDNA was explored by Isothermal Titration Calorimetry (ITC) and Differential Scanning Calorimetry (DSC). The ctDNA helix melting upon 6-Gingerol binding was examined by melting temperature Tm analysis. Further the electrophoretic mobility shift assay confirms a possible groove binding of 6-Gingerol with ctDNA. Molecular docking and Molecular dynamics (MD) studies provide a detailed understanding on the interaction of 6-Gingerol binding in the minor groove of DNA which supports experimental results.
Collapse
Affiliation(s)
- Poovvathingal Haris
- School of Pure and Applied Physics, Mahatma Gandhi University, Kottayam, Kerala 686560, India
| | - Varughese Mary
- School of Pure and Applied Physics, Mahatma Gandhi University, Kottayam, Kerala 686560, India
| | - Chellappanpillai Sudarsanakumar
- School of Pure and Applied Physics, Mahatma Gandhi University, Kottayam, Kerala 686560, India; Center for High Performance Computing, Mahatma Gandhi University, Kottayam, Kerala 686560, India.
| |
Collapse
|
33
|
Moosavi MA, Haghi A, Rahmati M, Taniguchi H, Mocan A, Echeverría J, Gupta VK, Tzvetkov NT, Atanasov AG. Phytochemicals as potent modulators of autophagy for cancer therapy. Cancer Lett 2018; 424:46-69. [PMID: 29474859 DOI: 10.1016/j.canlet.2018.02.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
The dysregulation of autophagy is involved in the pathogenesis of a broad range of diseases, and accordingly universal research efforts have focused on exploring novel compounds with autophagy-modulating properties. While a number of synthetic autophagy modulators have been identified as promising cancer therapy candidates, autophagy-modulating phytochemicals have also attracted attention as potential treatments with minimal side effects. In this review, we firstly highlight the importance of autophagy and its relevance in the pathogenesis and treatment of cancer. Subsequently, we present the data on common phytochemicals and their mechanism of action as autophagy modulators. Finally, we discuss the challenges associated with harnessing the autophagic potential of phytochemicals for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, P.O Box:14965/161, Tehran, Iran.
| | - Atousa Haghi
- Young Researchers & Elite Club, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Haţieganu" University of Medicine and Pharmacy, Gheorghe Marinescu 23 Street, 400337 Cluj-Napoca, Romania
| | - Javier Echeverría
- Facultad de Química y Biología, Universidad de Santiago de Chile, Casilla 40, Correo 33, Santiago 9170022, Chile
| | - Vijai K Gupta
- Department of Chemistry and Biotechnology, ERA Chair of Green Chemistry, Tallinn University of Technology, 12618 Tallinn, Estonia
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, Sofia 1618, Bulgaria
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
34
|
Mukkavilli R, Yang C, Tanwar RS, Saxena R, Gundala SR, Zhang Y, Ghareeb A, Floyd SD, Vangala S, Kuo WW, Rida PCG, Aneja R. Pharmacokinetic-pharmacodynamic correlations in the development of ginger extract as an anticancer agent. Sci Rep 2018; 8:3056. [PMID: 29445099 PMCID: PMC5813242 DOI: 10.1038/s41598-018-21125-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022] Open
Abstract
Anticancer efficacy of ginger phenolics (GPs) has been demonstrated in various in vitro assays and xenograft mouse models. However, only sub-therapeutic plasma concentrations of GPs were detected in human and mouse pharmacokinetic (PK) studies. Intriguingly, a significant portion of GPs occurred as phase II metabolites (mainly glucuronide conjugates) in plasma. To evaluate the disposition of GPs and understand the real players responsible for efficacy, we performed a PK and tissue distribution study in mice. Plasma exposure of GPs was similar on day 1 and 7, suggesting no induction or inhibition of clearance pathways. Both free and conjugated GPs accumulated in all tissues including tumors. While non-cytotoxicity of 6-ginerol glucuronide precluded the role of conjugated GPs in cell death, the free forms were cytotoxic against prostate cancer cells. The efficacy of ginger was best explained by the reconversion of conjugated GPs to free forms by β-glucuronidase, which is over-expressed in the tumor tissue. This previously unrecognized two-step process suggests an instantaneous conversion of ingested free GPs into conjugated forms, followed by their subsequent absorption into systemic circulation and reconversion into free forms. This proposed model uncovers the mechanistic underpinnings of ginger's anticancer activity despite sub-therapeutic levels of free GPs in the plasma.
Collapse
Affiliation(s)
- Rao Mukkavilli
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Chunhua Yang
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | | | - Roopali Saxena
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Sushma R Gundala
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Yingyi Zhang
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Ahmed Ghareeb
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Stephan D Floyd
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | | | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA.
| |
Collapse
|
35
|
Suvarna V, Murahari M, Khan T, Chaubey P, Sangave P. Phytochemicals and PI3K Inhibitors in Cancer-An Insight. Front Pharmacol 2017; 8:916. [PMID: 29311925 PMCID: PMC5736021 DOI: 10.3389/fphar.2017.00916] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/11/2022] Open
Abstract
In today's world of modern medicine and novel therapies, cancer still remains to be one of the prime contributor to the death of people worldwide. The modern therapies improve condition of cancer patients and are effective in early stages of cancer but the advanced metastasized stage of cancer remains untreatable. Also most of the cancer therapies are expensive and are associated with adverse side effects. Thus, considering the current status of cancer treatment there is scope to search for efficient therapies which are cost-effective and are associated with lesser and milder side effects. Phytochemicals have been utilized for many decades to prevent and cure various ailments and current evidences indicate use of phytochemicals as an effective treatment for cancer. Hyperactivation of phosphoinositide 3-kinase (PI3K) signaling cascades is a common phenomenon in most types of cancers. Thus, natural substances targeting PI3K pathway can be of great therapeutic potential in the treatment of cancer patients. This chapter summarizes the updated research on plant-derived substances targeting PI3K pathway and the current status of their preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Vasanti Suvarna
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Manikanta Murahari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S Ramaiah University of Applied Sciences, Bangalore, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pramila Chaubey
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Preeti Sangave
- Department of Pharmaceutical Sciences, School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, India
| |
Collapse
|
36
|
Iqbal J, Abbasi BA, Mahmood T, Kanwal S, Ali B, Shah SA, Khalil AT. Plant-derived anticancer agents: A green anticancer approach. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.10.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
37
|
Wang Q, Wei Q, Yang Q, Cao X, Li Q, Shi F, Tong SS, Feng C, Yu Q, Yu J, Xu X. A novel formulation of [6]-gingerol: Proliposomes with enhanced oral bioavailability and antitumor effect. Int J Pharm 2017; 535:308-315. [PMID: 29126908 DOI: 10.1016/j.ijpharm.2017.11.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/13/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023]
Abstract
[6]-Gingerol, one of the components of the rhizome of Ginger, has a variety of biological activities such as anticoagulant, antioxidative, antitumor, anti-inflammatory, antihypertensive, and so forth. However, as one of the homologous phenolic ketones, [6]-gingerol is insoluble in water which limits its applications. Herein, we prepared [6]-gingerol proliposomes through modified thin-film dispersion method, which was spherical or oval, and physicochemically stable with narrow size distribution. Surprisingly, in vitro release of [6]-gingerol loaded proliposome compared with the free [6]-gingerol was significantly higher and its oral bioavailability increased 5-fold in vivo. Intriguingly, its antitumor effect was enhanced in the liposome formulation. Thus, our prepared [6]-gingerol proliposome proved to be a novel formulation for [6]-gingerol, which significantly improved its antitumor effect.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Qiuyu Wei
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Qiuxuan Yang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Qiang Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Feng Shi
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Shan Shan Tong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Chunlai Feng
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Qingtong Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China.
| |
Collapse
|
38
|
Zhang F, Zhang JG, Qu J, Zhang Q, Prasad C, Wei ZJ. Assessment of anti-cancerous potential of 6-gingerol (Tongling White Ginger) and its synergy with drugs on human cervical adenocarcinoma cells. Food Chem Toxicol 2017; 109:910-922. [PMID: 28249781 DOI: 10.1016/j.fct.2017.02.038] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 12/11/2022]
Abstract
The anti-cancerous activity of 6-gingerol extracted from Tongling White Ginger was investigated. 6-Gingerol inhibited the growth of HeLa cells with IC50 (96.32 μM) and IC80 (133.01 μM) and led to morphological changes, induced the cell cycle arrest in G0/G1-phase and ultimately resulted into apoptosis. Among cell cycle-related genes and proteins, the expression of cyclin (A, D1, E1) reduced, while of CDK-1, p21 and p27 showed slight decrease, except cyclin B1 and E1 (protein). Western blotting reported the induction of apoptosis with an increased Bax/Bcl-2 ratio, release of cytochrome c, cleavage of caspase-3, -8, -9 and PRPP in treated cells. 6-Gingerol activated AMPK, but inhibited PI3K/AKT phosphorylation with reduced P70S6K expression and also suppressed the mTOR phosphorylation. 6-Gingerol with 5-FU and Ptx resulted in 83.2% and 52% inhibition respectively, this synergy have stimulated apoptosis proteins more efficiently as compared to 6-Gingerol alone (10.75%) under in vitro conditions.
Collapse
Affiliation(s)
- Fang Zhang
- School of Food Science and Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Jian-Guo Zhang
- School of Food Science and Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Jie Qu
- School of Food Science and Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Qi Zhang
- School of Food Science and Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Chandan Prasad
- Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX, USA.
| | - Zhao-Jun Wei
- School of Food Science and Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| |
Collapse
|
39
|
Mohamed SIA, Jantan I, Haque MA. Naturally occurring immunomodulators with antitumor activity: An insight on their mechanisms of action. Int Immunopharmacol 2017; 50:291-304. [PMID: 28734166 DOI: 10.1016/j.intimp.2017.07.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/13/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
|
40
|
Fuzer AM, Lee SY, Mott JD, Cominetti MR. [10]-Gingerol Reverts Malignant Phenotype of Breast Cancer Cells in 3D Culture. J Cell Biochem 2017; 118:2693-2699. [PMID: 28112417 DOI: 10.1002/jcb.25906] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/20/2017] [Indexed: 01/04/2023]
Abstract
Breast cancer is a complex and multifactorial disease. Tumors have a heterogeneous microenvironment, which have multiple interactions with other cell types, greatly influencing the behavior of tumor cells and response to therapy. The 3D culture mimics the microenvironment better found in vivo and is more appropriated than the traditional 2D culture made from plastic to test the cellular response to drugs. To investigate the effects of [10]-gingerol on breast tumor cells, we used physiologically relevant three-dimensional (3D) cultures of malignant and non-malignant human breast cells grown in laminin-rich extracellular matrix gels (lr-ECM). Our results showed selective cytotoxicity of [10]-gingerol against the malignant T4-2 breast cancer cell line compared to non-malignant S1 cells. The compound reverted the malignant phenotype of the cancer cells, downregulating the expression of epidermal growth factor receptor (EGFR) and β1-integrin. Moreover, [10]-gingerol induced apoptosis in this cell line. These results suggest that [10]-gingerol may be an effective compound to use as adjuvant therapy in breast cancer treatment. J. Cell. Biochem. 118: 2693-2699, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Angelina M Fuzer
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Sun-Young Lee
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Joni D Mott
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil
| |
Collapse
|
41
|
Švarc-Gajić J, Cvetanović A, Segura-Carretero A, Linares IB, Mašković P. Characterisation of ginger extracts obtained by subcritical water. J Supercrit Fluids 2017. [DOI: 10.1016/j.supflu.2016.12.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
42
|
Zhang F, Thakur K, Hu F, Zhang JG, Wei ZJ. 10-Gingerol, a Phytochemical Derivative from "Tongling White Ginger", Inhibits Cervical Cancer: Insights into the Molecular Mechanism and Inhibitory Targets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:2089-2099. [PMID: 28230361 DOI: 10.1021/acs.jafc.7b00095] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
With the aim of evaluating anticancerous activities of 10-gingerol (10-G) against HeLa cells, it was purified and identified from "Tongling white ginger" by HSCCC, UPLC-TOF-MS/MS, and NMR analysis, respectively. 10-G inhibited the proliferation of HeLa cells at IC50 (29.19 μM) and IC80 (50.87 μM) with altered cell morphology, increased cytotoxicity, and arrested cell cycle in the G0/G1 phase. Most cell cycle related genes and protein expression significantly decreased, followed by a slight decrease in a few without affecting cyclin B1 and cyclin E1 (protein). Both death receptors significantly up-regulated and activated apoptosis indicators (caspase family). Furthermore, significant changes in mitochondria-dependent pathway markers were observed and led to cell death. 10-G led to PI3K/AKT inhibition and AMPK activation to induce mTOR-mediated cell apoptosis in HeLa cells. These results can be an asset to exploit 10-G with other medicinal plant derivatives for future applications.
Collapse
Affiliation(s)
- Fang Zhang
- School of Food Science and Engineering, Hefei University of Technology , Hefei 230009, People's Republic of China
| | - Kiran Thakur
- School of Food Science and Engineering, Hefei University of Technology , Hefei 230009, People's Republic of China
| | - Fei Hu
- School of Food Science and Engineering, Hefei University of Technology , Hefei 230009, People's Republic of China
| | - Jian-Guo Zhang
- School of Food Science and Engineering, Hefei University of Technology , Hefei 230009, People's Republic of China
| | - Zhao-Jun Wei
- School of Food Science and Engineering, Hefei University of Technology , Hefei 230009, People's Republic of China
- Agricultural and Forestry Specialty Food Processing Industry Technological Innovation Strategic Alliance of Anhui Province , Hefei 230009, People's Republic of China
| |
Collapse
|
43
|
Abstract
Cancer remains one of the leading causes of death around the world. Initially it is recognized as a genetic disease, but now it is known to involve epigenetic abnormalities along with genetic alterations. Epigenetics refers to heritable changes that are not encoded in the DNA sequence itself, but play an important role in the control of gene expression. It includes changes in DNA methylation, histone modifications, and RNA interference. Although it is heritable, environmental factors such as diet could directly influence epigenetic mechanisms in humans. This article will focus on the role of dietary patterns and phytochemicals that have been demonstrated to influence the epigenome and more precisely histone and non-histone proteins modulation by acetylation that helps to induce apoptosis and phosphorylation inhibition, which counteracts with cells proliferation. Recent developments discussed here enhance our understanding of how dietary intervention could be beneficial in preventing or treating cancer and improving health outcomes.
Collapse
Affiliation(s)
- Wissam Zam
- a Department of Analytical and Food Chemistry , Faculty of Pharmacy, Al-Andalus University for Medical Sciences, Al-Quadmous , Tartous , Syrian Arab Republic
| | - Aziz Khadour
- b Department of Microbiology , Faculty of Pharmacy, Al-Andalus University for Medical Sciences, Al-Quadmous , Tartous , Syrian Arab Republic
| |
Collapse
|
44
|
Singh P, Arora D, Shukla Y. Enhanced chemoprevention by the combined treatment of pterostilbene and lupeol in B[a]P-induced mouse skin tumorigenesis. Food Chem Toxicol 2017; 99:182-189. [DOI: 10.1016/j.fct.2016.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 12/16/2022]
|
45
|
Rastogi N, Duggal S, Singh SK, Porwal K, Srivastava VK, Maurya R, Bhatt MLB, Mishra DP. Proteasome inhibition mediates p53 reactivation and anti-cancer activity of 6-gingerol in cervical cancer cells. Oncotarget 2016; 6:43310-25. [PMID: 26621832 PMCID: PMC4791234 DOI: 10.18632/oncotarget.6383] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/17/2015] [Indexed: 12/15/2022] Open
Abstract
Human papilloma virus (HPV) expressing E6 and E7 oncoproteins, is known to inactivate the tumor suppressor p53 through proteasomal degradation in cervical cancers. Therefore, use of small molecules for inhibition of proteasome function and induction of p53 reactivation is a promising strategy for induction of apoptosis in cervical cancer cells. The polyphenolic alkanone, 6-Gingerol (6G), present in the pungent extracts of ginger (Zingiber officinale Roscoe) has shown potent anti-tumorigenic and pro-apoptotic activities against a variety of cancers. In this study we explored the molecular mechanism of action of 6G in human cervical cancer cells in vitro and in vivo. 6G potently inhibited proliferation of the HPV positive cervical cancer cells. 6G was found to: (i) inhibit the chymotrypsin activity of proteasomes, (ii) induce reactivation of p53, (iii) increase levels of p21, (iv) induce DNA damage and G2/M cell cycle arrest, (v) alter expression levels of p53-associated apoptotic markers like, cleaved caspase-3 and PARP, and (vi) potentiate the cytotoxicity of cisplatin. 6G treatment induced significant reduction of tumor volume, tumor weight, proteasome inhibition and p53 accumulation in HeLa xenograft tumor cells in vivo. The 6G treatment was devoid of toxic effects as it did not affect body weights, hematological and osteogenic parameters. Taken together, our data underscores the therapeutic and chemosensitizing effects of 6G in the management and treatment of cervical cancer.
Collapse
Affiliation(s)
- Namrata Rastogi
- Cell Death Research Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shivali Duggal
- Department of Radiotherapy, King George Medical University, Lucknow, India
| | - Shailendra Kumar Singh
- Department of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Konica Porwal
- Cell Death Research Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | | | - Rakesh Maurya
- Medicinal Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - M L B Bhatt
- Department of Radiotherapy, King George Medical University, Lucknow, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
46
|
Saha P, Katarkar A, Das B, Bhattacharyya A, Chaudhuri K. 6-Gingerol inhibits Vibrio cholerae-induced proinflammatory cytokines in intestinal epithelial cells via modulation of NF-κB. PHARMACEUTICAL BIOLOGY 2016; 54:1606-1615. [PMID: 26987371 DOI: 10.3109/13880209.2015.1110598] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/28/2015] [Accepted: 10/14/2015] [Indexed: 06/05/2023]
Abstract
Context The effect of 6-gingerol (6G), the bioactive component of Zingiber officinale Roscoe (Zingiberaceae), in the reduction of Vibrio cholerae (Vibrionaceae)-induced inflammation has not yet been reported. Materials and methods Cell viability assay was performed to determine the working concentration of 6G. Elisa and RT-PCR were performed with Int 407 cells treated with 50 μM 6G and 100 multiplicity of infection (MOI) V. cholerae for 0, 2, 3, 3.5, 6 and 8 h to determine the concentration of IL-8, IL-6, IL-1α and IL-1β in both protein and RNA levels. Furthermore, the effect of 50 μM 6G on upstream MAP-kinases and NF-κB signalling pathways was evaluated at 0, 10, 15, 30, 60 and 90 min. Results The effective dose (ED50) value of 6G was found to be 50 μM as determined by cell viability assay. Pre-treatment with 50 μM 6G reduced V. cholerae infection-triggered levels of IL-8, IL-6, IL-1α and IL-1β by 3.2-fold in the protein level and two-fold in the RNA level at 3.5 h. The levels of MAP-kinases signalling molecules like p38 and ERK1/2 were also reduced by two- and three-fold, respectively, after 30 min of treatment. Additionally, there was an increase in phosphorylated IκBα and down-regulation of p65 resulting in down-regulation of NF-κB pathway. Conclusion Our results showed that 6G could modulate the anti-inflammatory responses triggered by V. cholerae-induced infection in intestinal epithelial cells by modulating NF-κB pathway.
Collapse
Affiliation(s)
- Pallashri Saha
- a Molecular and Human Genetics Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Atul Katarkar
- a Molecular and Human Genetics Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Bornita Das
- a Molecular and Human Genetics Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Aritra Bhattacharyya
- a Molecular and Human Genetics Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Keya Chaudhuri
- a Molecular and Human Genetics Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India
| |
Collapse
|
47
|
TIWARI MANISHK, MISHRA PC. Anti-oxidant activity of 6-gingerol as a hydroxyl radical scavenger by hydrogen atom transfer, radical addition and electron transfer mechanisms. J CHEM SCI 2016. [DOI: 10.1007/s12039-016-1128-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
48
|
Sirdaarta J, Maen A, Rayan P, Matthews B, Cock IE. High Performance Liquid Chromatography-mass Spectrometry Analysis of High Antioxidant Australian Fruits with Antiproliferative Activity Against Cancer Cells. Pharmacogn Mag 2016; 12:S181-94. [PMID: 27279705 PMCID: PMC4883077 DOI: 10.4103/0973-1296.182178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 07/01/2015] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND High antioxidant capacities have been linked to the treatment and prevention of several cancers. Recent reports have identified several native Australian fruits with high antioxidant capacities. Despite this, several of these species are yet to be tested for anticancer activity. MATERIALS AND METHODS Solvent extracts prepared from high antioxidant native Australian fruits were analyzed for antioxidant capacity by the di (phenyl)-(2,4,6-trinitrophenyl) iminoazanium free radical scavenging assay. Antiproliferative activities against CaCo2 and HeLa cancer cells were determined by a multicellular tumor spheroid-based cell proliferation assay. Toxicity was determined by Artemia franciscana bioassay. RESULTS Methanolic extracts of all plant species displayed high antioxidant contents (equivalent to approximately 7-16 mg of vitamin C per gram of fruit extracted). Most aqueous extracts also contained relatively high antioxidant capacities. In contrast, the ethyl acetate, chloroform, and hexane extracts of most species (except lemon aspen and bush tomato) had lower antioxidant contents (below 1.5 mg of vitamin C equivalents per gram of plant material extracted). The antioxidant contents correlated with the ability of the extracts to inhibit proliferation of CaCo2 and HeLa cancer cell lines. The high antioxidant methanolic extracts of all species were potent inhibitors of cell proliferation. The methanolic lemon aspen extract was particularly effective, with IC50 values of 480 and 769 μg/mL against HeLa and CaCo2 cells, respectively. In contrast, the lower antioxidant ethyl acetate and hexane extracts (except the lemon aspen ethyl acetate extract) generally did not inhibit cancer cell proliferation or inhibited to only a minor degree. Indeed, most of the ethyl acetate and hexane extracts induced potent cell proliferation. The native tamarind ethyl acetate extract displayed low-moderate toxicity in the A. franciscana bioassay (LC50 values below 1000 μg/mL). All other extracts were nontoxic. A total of 145 unique mass signals were detected in the lemon aspen methanolic and aqueous extracts by nonbiased high-performance liquid chromatography-mass spectrometry analysis. Of these, 20 compounds were identified as being of particular interest due to their reported antioxidant and/or anticancer activities. CONCLUSIONS The lack of toxicity and antiproliferative activity of the high antioxidant plant extracts against HeLa and CaCo2 cancer cell lines indicates their potential in the treatment and prevention of some cancers. SUMMARY Australian fruit extracts with high antioxidant contents were potent inhibitors of CaCo2 and HeLa carcinoma cell proliferationMethanolic lemon aspen extract was particularly potent, with IC50 values of 480 μg/mL (HeLa) and 769 μg/mL (CaCo2)High-performance liquid chromatography-mass spectrometry-quadrupole time-of-flight analysis highlighted and putatively identified 20 compounds in the antiproliferative lemon aspen extractsIn contrast, lower antioxidant content extracts stimulated carcinoma cell proliferationAll extracts with antiproliferative activity were nontoxic in the Artemia nauplii assay. Abbreviations used: DPPH: di (phenyl)- (2,4,6-trinitrophenyl) iminoazanium, HPLC: High-performance liquid chromatography, IC50: The concentration required to inhibit by 50%, LC50: The concentration required to achieve 50% mortality, MS: Mass spectrometry.
Collapse
Affiliation(s)
- Joseph Sirdaarta
- Environmental Futures Research Institute, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia; School of Natural Sciences, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia
| | - Anton Maen
- School of Natural Sciences, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia
| | - Paran Rayan
- Environmental Futures Research Institute, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia; School of Natural Sciences, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia
| | - Ben Matthews
- Smart Water Research Centre, Griffith University, Gold Coast, 4222 Queensland, Australia
| | - Ian Edwin Cock
- Environmental Futures Research Institute, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia; School of Natural Sciences, Nathan Campus, Griffith University, Nathan, 4111 Queensland, Australia
| |
Collapse
|
49
|
Kapoor V, Aggarwal S, Das SN. 6-Gingerol Mediates its Anti Tumor Activities in Human Oral and Cervical Cancer Cell Lines through Apoptosis and Cell Cycle Arrest. Phytother Res 2016; 30:588-95. [PMID: 26749462 DOI: 10.1002/ptr.5561] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 11/26/2015] [Accepted: 12/04/2015] [Indexed: 11/08/2022]
Abstract
6-Gingerol, a potent nutraceutical, has been shown to have antitumor activity in different tumors, although its mechanism of action is not well understood. In this study, we evaluated antitumor activities of 6-gingerol on human oral (SCC4, KB) and cervical cancer (HeLa) cell lines with or without wortmannin, rapamycin, and cisplatin. Tumor cell proliferation was observed using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H tetrazolium, inner salt assay, cell cycle analysis by propidium iodide labeling and flow cytometry, apoptosis by Annexin-V binding assay, and caspase activity by chemiluminescence assay. 6-Gingerol showed dose-dependent cytotoxicity in all three cell lines. Combinations of 6-gingerol with wortmannin and cisplatin showed additive effects, while with rapamycin, it showed 50% cytotoxicity that was equivalent to IC50 of 6-gingerol alone. Treatment with 6-gingerol resulted in G2-phase arrest in KB and HeLa cells and S-phase arrest in SCC4 cells. 6-Gingerol, wortmannin, and rapamycin treatment showed almost two-fold higher expression of caspase 3 in all cell lines. The results imply that 6-gingerol either alone or in combination with PI-3 K inhibitor and cisplatin may provide better therapeutic effects in oral and cervical carcinoma. Thus, 6-gingerol appears to be a safe and potent chemotherapeutic/chemopreventive compound acting through cell cycle arrest and induction of apoptosis in human oral and cervical tumor cells.
Collapse
Affiliation(s)
- Vaishali Kapoor
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sadhna Aggarwal
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Satya N Das
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
50
|
Mohd Yusof YA. Gingerol and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 929:177-207. [PMID: 27771925 DOI: 10.1007/978-3-319-41342-6_8] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Since antiquity, ginger or Zingiber officinale, has been used by humans for medicinal purposes and as spice condiments to enhance flavor in cooking. Ginger contains many phenolic compounds such as gingerol, shogaol and paradol that exhibit antioxidant, anti-tumor and anti-inflammatory properties. The role of ginger and its constituents in ameliorating diseases has been the focus of study in the past two decades by many researchers who provide strong scientific evidence of its health benefit. This review discusses research findings and works devoted to gingerols, the major pungent constituent of ginger, in modulating and targeting signaling pathways with subsequent changes that ameliorate, reverse or prevent chronic diseases in human studies and animal models. The physical, chemical and biological properties of gingerols are also described. The use of ginger and especially gingerols as medicinal food derivative appears to be safe in treating or preventing chronic diseases which will benefit the common population, clinicians, patients, researchers, students and industrialists.
Collapse
Affiliation(s)
- Yasmin Anum Mohd Yusof
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Level 17, Pre-Clinical Building, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|