1
|
Sirizi MAG, Alizadeh Ghalenoei J, Allahtavakoli M, Forouzanfar H, Bagheri SM. Anticancer potential of Ferula assa-foetida and its constituents, a powerful plant for cancer therapy. World J Biol Chem 2023; 14:28-39. [PMID: 37034135 PMCID: PMC10080545 DOI: 10.4331/wjbc.v14.i2.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/24/2023] Open
Abstract
Cancer is one of the main challenges of the health system around the world. This disease is increasing in developing countries and imposes heavy costs on patients and governments. On the other hand, despite various drugs, the death rate among cancer patients is still high and the current treatments have many harmful effects. In the traditional medicine of different countries, there are many medicinal plants that can be effective in the treatment of cancer. Ferula plants are traditionally used as spices and food or for medicinal purposes. Ferula assa-foetida is one of the famous plants of this genus, which has been used for the treatment of various diseases since ancient times. Among the main compounds of this plant, we can mention monoterpenes, sulfide compounds and polyphenols, which can show different therapeutic effects. This article has been compiled with the aim of collecting evidence and articles related to the anti-cancer effects of extracts, derived compounds, essential oils and nanoparticles containing Ferula assa-foetida. This review article was prepared by searching the terms Ferula assa-foetida and cancer, and relevant information was collected through searching electronic databases such as ISI Web of Knowledge, PubMed, and Google Scholar. Fortunately, the results of this review showed that relatively comprehensive studies have been conducted in this field and shown that Ferula assa-foetida can be very promising in the treatment of cancer.
Collapse
Affiliation(s)
- Mohammad Amin Ghaffari Sirizi
- Department of Physiology, Hematology-oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd 8915173149, Iran
| | - Jalil Alizadeh Ghalenoei
- Department of Physiology, Hematology-oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd 8915173149, Iran
| | - Mohammad Allahtavakoli
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 8915173149, Iran
| | - Hasan Forouzanfar
- Department of Nursing, Tabas School of nursing, Birjand University of Medical Sciences, Birjand 8915173149, Iran
| | - Seyyed Majid Bagheri
- Department of Physiology, Hematology-oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd 8915173149, Iran
| |
Collapse
|
2
|
Li X, Zhang Y, Wang N, Yuan Z, Chen X, Chen Q, Deng H, Tong X, Chen H, Duan Y, Wei Y. CircRNA.0007127 triggers apoptosis through the miR-513a-5p/CASP8 axis in K-562 cells. J Zhejiang Univ Sci B 2022; 23:732-746. [PMID: 36111570 DOI: 10.1631/jzus.b2200048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND: Circular RNAs (circRNAs) are covalently closed single-stranded RNAs with multiple biological functions. CircRNA.0007127 is derived from the carbon catabolite repression 4-negative on TATA-less (CCR4-NOT) complex subunit 2 (CNOT2), which was found to regulate tumor cell apoptosis through caspase pathway. METHODS: Potential circRNA.0007127 target microRNAs (miRNAs) were analyzed by miRanda, TargetScan, and RNAhybrid software, and the miRNAs with binding sites for apoptosis-related genes were screened. The roles of circRNA.0007127 and its downstream target, microRNA (miR)-513a-5p, were validated by quantitative real-time polymerase chain reaction (qPCR), flow cytometry, mitochondrial membrane potential, immunofluorescence, western blot, and caspase-8 (CASP8) protein activity in vitro in H2O2-induced K-562 cells. The circRNA.0007127‒miR-513a-5p and CASP8‒miR-513a-5p interactions were verified by luciferase reporter assays. RESULTS: Silencing circRNA.0007127 decreased cell apoptosis by inhibiting CASP8 pathway activation in K-562 cells. Compared with the control group, the expression of CASP8 was reduced by 50% and the 43-kD fragment of CASP8 protein was significantly reduced (P≤0.05). The luciferase reporting assay showed that circRNA.0007127 combined with miR-513a-5p or CASP8, with extremely significant differences (P≤0.001). The overexpression of miR-513a-5p inhibited the gene expression level of CASP8 in a human myeloid leukemia cell model (75% change) and the level of a 43-kD fragment of CASP8 protein (P≤0.01). The rescue experiment showed that cotransfection with circRNA.0007127 small-interfering RNA (siRNA) and the miR-513a-5p inhibitor increased CASP8 gene expression and the apoptosis rate, suggesting that the miR-513a-5p inhibitor is a circRNA.0007127 siRNA antagonist. CONCLUSIONS: CircRNA.0007127 regulates K-562 cell apoptosis through the miR-513a-5p/CASP8 axis, which can serve as a novel powerful molecular target for K-562 cells.
Collapse
Affiliation(s)
- Xiajing Li
- School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Yiyu Zhang
- Department of Blood Transfusion, Shenzhen Longhua Central Hospital, Shenzhen 518000, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510000, China.,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510000, China
| | - Zhaohu Yuan
- Department of Blood Transfusion, the Second Affiliation Hospital of South China University of Technology, Guangzhou 510000, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou 510000, China
| | - Xiaojie Chen
- Department of Blood Transfusion, the Second Affiliation Hospital of South China University of Technology, Guangzhou 510000, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou 510000, China
| | - Qicong Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510000, China.,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510000, China
| | - Hui Deng
- Department of Blood Transfusion, the Second Affiliation Hospital of South China University of Technology, Guangzhou 510000, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou 510000, China
| | - Xinxin Tong
- Department of Blood Transfusion, the Second Affiliation Hospital of South China University of Technology, Guangzhou 510000, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou 510000, China
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510000, China. ,
| | - Yaming Wei
- Department of Blood Transfusion, the Second Affiliation Hospital of South China University of Technology, Guangzhou 510000, China. .,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou 510000, China.
| |
Collapse
|
3
|
Punpai S, Saenkham A, Jarintanan F, Jongrungruangchok S, Choowongkomon K, Suksamrarn S, Tanechpongtamb W. HDAC inhibitor cowanin extracted from G. fusca induces apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathways in Jurkat cells. Biomed Pharmacother 2022; 147:112577. [PMID: 35078092 DOI: 10.1016/j.biopha.2021.112577] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Cowanin, a xanthone derivative extracted from the Garcinia fusca plant, has been recognized for various biological activities including, antimicrobial, anti-inflammatory, and anticancer activities. However, the mechanism to induce cancer cell death in cancer cells remains to be fully elucidated. Our previous report showed that other xanthones from these plants could act as histone deacetylase inhibitors (HDACi), so we deeply analyzed the role of cowanin, a major compound of G.fusca, and investigated through the mode of cell death both apoptosis and autophagy that have never been reported. As a result, it was demonstrated that cowanin indicated the role of HDACi as other xanthones. The molecular docking analysis showed that cowanin could interact within the catalytic pocket region of HDAC class I (HDAC2, 8) and II (HDAC4, 7) proteins and inhibit their activity. Also, the level of protein expression of HDAC2, 4, 7, and 8 was distinctly decreased, and the level of histone H3 and H4 acetylation increased in cowanin treated cells. For the mode of cell death, cowanin demonstrated both apoptosis and autophagy activation in Jurkat cells. Besides, cowanin significantly suppressed phosphorylation of PI3K, Akt, and mTOR signaling. Therefore, these findings revealed that cowanin represents a new promising candidate for development as an anticancer agent by inducing apoptosis and autophagy via PI3K/AKT/mTOR pathway and effectively inhibiting HDAC activity.
Collapse
Affiliation(s)
- Sakdiphong Punpai
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Audchara Saenkham
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok 10110, Thailand
| | | | | | - Kiattawee Choowongkomon
- Departmentof Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10903, Thailand
| | - Sunit Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Wanlaya Tanechpongtamb
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand.
| |
Collapse
|