1
|
Wen C, Dechsupa N, Yu Z, Zhang X, Liang S, Lei X, Xu T, Gao X, Hu Q, Innuan P, Kantapan J, Lü M. Pentagalloyl Glucose: A Review of Anticancer Properties, Molecular Targets, Mechanisms of Action, Pharmacokinetics, and Safety Profile. Molecules 2023; 28:4856. [PMID: 37375411 DOI: 10.3390/molecules28124856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Pentagalloyl glucose (PGG) is a natural hydrolyzable gallotannin abundant in various plants and herbs. It has a broad range of biological activities, specifically anticancer activities, and numerous molecular targets. Despite multiple studies available on the pharmacological action of PGG, the molecular mechanisms underlying the anticancer effects of PGG are unclear. Here, we have critically reviewed the natural sources of PGG, its anticancer properties, and underlying mechanisms of action. We found that multiple natural sources of PGG are available, and the existing production technology is sufficient to produce large quantities of the required product. Three plants (or their parts) with maximum PGG content were Rhus chinensis Mill, Bouea macrophylla seed, and Mangifera indica kernel. PGG acts on multiple molecular targets and signaling pathways associated with the hallmarks of cancer to inhibit growth, angiogenesis, and metastasis of several cancers. Moreover, PGG can enhance the efficacy of chemotherapy and radiotherapy by modulating various cancer-associated pathways. Therefore, PGG can be used for treating different human cancers; nevertheless, the data on the pharmacokinetics and safety profile of PGG are limited, and further studies are essential to define the clinical use of PGG in cancer therapies.
Collapse
Affiliation(s)
- Chengli Wen
- Department of Intensive Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Luzhou Key Laboratory of Human Microecology and Precision Diagnosis and Treatment, Luzhou 646000, China
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Zehui Yu
- Laboratory Animal Center, Southwest Medical University, Luzhou 646000, China
| | - Xu Zhang
- Luzhou Key Laboratory of Human Microecology and Precision Diagnosis and Treatment, Luzhou 646000, China
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Sicheng Liang
- Luzhou Key Laboratory of Human Microecology and Precision Diagnosis and Treatment, Luzhou 646000, China
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xianying Lei
- Department of Intensive Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tao Xu
- Department of Intensive Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiaolan Gao
- Department of Intensive Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qinxue Hu
- Department of Intensive Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Muhan Lü
- Luzhou Key Laboratory of Human Microecology and Precision Diagnosis and Treatment, Luzhou 646000, China
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
2
|
Liu S, Cao X, Zhang T, Zhang C, Qu J, Sun Y, Lv W, Qu F. Paeonol ameliorates endometrial hyperplasia in mice via inhibiting PI3K/AKT pathway-related ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154593. [PMID: 36610113 DOI: 10.1016/j.phymed.2022.154593] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/24/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Paeonol (Pae) is one of the active ingredients from components of Guizhi Fuling Capsule, a traditional Chinese medicine widely used for the treatment of women's diseases, which exhibits various biological and pharmacological activities. PURPOSE The objective of this study was to investigate the molecular mechanism underlying the role of Pae in protecting against endometrial hyperplasia (EH). METHODS CCK-8 assay was performed to detect the effect of Pae on cell proliferation. Hematoxylin and eosin (H&E) staining was performed to evaluate uterine tissue structure. A network pharmacology study was performed to search the disease targets. Single-cell transcriptome analysis was performed with uterine tissues from 3 healthy donors and 3 EH patients on 10X Genomics platform. Changes in lipid peroxidation were detected by the MDA reaction. IHC assay, Western blot, immunofluorescence and RT-qPCR were used to study the effects of estradiol and Pae on the expression levels of GPX4, PI3K, AKT, p-PI3K, p-AKT in mice. RESULTS Pae treatment resulted in a decrease in cell viability of endometrial epithelial cells. Loss of uterus weight and morphology changes were observed in mice. In addition, Fe iron concentration and MDA levels increased, while the expression of GPX4, p-PI3K and p-AKT diminished. CONCLUSIONS Pae exhibited obvious alleviative activity in estradiol-induced mice via PI3K/AKT signaling pathway-regulated ferroptosis.
Collapse
Affiliation(s)
- Songjun Liu
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Hangzhou 310006, Zhejiang, China; Department of Gynecology, Tongde Hospital of Zhejiang Province, 234 Gucui Road, Hangzhou 310012, Zhejiang, China
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China.
| | - Wen Lv
- Department of Gynecology, Tongde Hospital of Zhejiang Province, 234 Gucui Road, Hangzhou 310012, Zhejiang, China.
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
3
|
Liu CJ, Fan XD, Jiang JG, Chen QX, Zhu W. Potential anticancer activities of securinine and its molecular targets. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154417. [PMID: 36063584 DOI: 10.1016/j.phymed.2022.154417] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Securinine is an alkaloid identified from the roots and leaves of the shrub Flueggea suffruticosa (Pall.) Baill. The molecular structure of securinine consists of four rings, including three chiral centers. It has been suggested that securinine can be chemically synthesized from tyrosine and lysine. Securinine has long been used to treat central nervous system diseases. In recent years, more and more evidence shows that securinine also has anticancer activity, which has not been systematically discussed and analyzed. PURPOSE This study aims to propose an overall framework to describe the molecular targets of securinine in different signal pathways, and discuss the current status and prospects of each pathway, so as to provide a theoretical basis for the development securinine as an effective anticancer drug. METHODS The research databases on the anticancer activity of securinine from PubMed, Scopus, Web of Science and ScienceDirect to 2021 were systematically searched. This paper follows the Preferred Reporting Items and Meta-Analysis guidelines. RESULTS Securinine has the ability to kill a variety of human cancer cells, including, leukemia as well as prostate, cervical, breast, lung, and colon cancer cells. It can regulate the signal pathways of phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin, Wnt and Janus kinase-signal transducer and activator of transcription, promote cancer cell apoptosis and autophagy, and inhibit cancer cell metastasis. Securinine also has the activity of inducing leukemia cell differentiation. CONCLUSION Although there has been some experimental evidence indicating the anticancer effect of securinine and its possible pharmacology, in order to design more effective anticancer drugs, it is necessary to study the synergy of intracellular signaling pathways. More in vivo experiments and even clinical studies are needed, and the synergy between securinine and other drugs is also worth studying.
Collapse
Affiliation(s)
- Chang-Jun Liu
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Xiao-Dan Fan
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Jian-Guo Jiang
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China.
| | - Qiu-Xiong Chen
- The second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Wei Zhu
- The second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| |
Collapse
|
4
|
Yang H, Yue GGL, Leung PC, Wong CK, Zhang YJ, Lau CBS. Anti-metastatic effects of 1,2,3,4,6-Penta-O-galloyl-β-D-glucose in colorectal cancer: Regulation of cathepsin B-mediated extracellular matrix dynamics and epithelial-to-mesenchymal transition. Pharmacol Res 2022; 184:106457. [PMID: 36116708 DOI: 10.1016/j.phrs.2022.106457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 02/05/2023]
Abstract
Despite significant advances in the diagnosis and treatment of colorectal cancer (CRC), metastatic colorectal cancer still poses serious threat to CRC patients. The natural gallotannin 1,2,3,4,6-penta-O-galloyl-β-D-glucose (PGG) has been shown to possess anti-tumor effects on colon cancer cells, but its anti-metastatic effect is yet to be investigated. In this study, the effects of PGG on cell proliferation, colony formation ability, motility, migration were investigated in colon cancer cells using BrdU, colony formation, scratch, and transwell assays, respectively. Western blot assay was used for assessing protein expression. The orthotopic colon tumor-bearing mouse model and human colon cancer metastatic mouse model were employed to evaluate the anti-metastatic effects of PGG. Results showed that PGG exhibited not only anti-proliferative and colony formation inhibitory effects, but also inhibition on cell adhesion, motility, and migration in both HCT116 and colon 26-M01 cells via modulating protein expression of cathepsin B, FAK, cofilin, and epithelial-to-mesenchymal transition related proteins. In addition, PGG (10 or 15 mg/kg, i.p.) could significantly inhibit liver and lung metastasis in colon cancer metastatic mice models. Furthermore, PGG could regulate the populations of T cells, macrophages, and MDSCs, while the levels of IL-2, IL-6, IL-10, IFN-γ, and TNF-α were altered after PGG treatment in metastatic CRC mice. This is the first report of the anti-metastatic effects of PGG by regulating cathepsin B-mediated extracellular matrix dynamics and epithelial-to-mesenchymal transition process in CRC. Our findings suggested that PGG has great potential to be developed as an anti-metastatic agent for metastatic CRC.
Collapse
Affiliation(s)
- Huihai Yang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ping-Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Chun-Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China; Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ying-Jun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, People's Republic of China.
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China.
| |
Collapse
|
5
|
Wan H, Feng Y, Wu J, Zhu L, Mi Y. Functions and mechanisms of N6‑methyladenosine in prostate cancer (Review). Mol Med Rep 2022; 26:280. [PMID: 35856412 PMCID: PMC9364137 DOI: 10.3892/mmr.2022.12796] [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: 05/23/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (PCa) has long been a major public health problem affecting men worldwide. Even with treatment, it can develop into castration-resistant PCa. With the continuous advancement in epigenetics, researchers have explored N6-methyladenosine (m6A) in search of a more effective and lasting treatment for PCa. m6A is widely distributed in mammalian cells and influences various aspects of mRNA metabolism. Recently, it has been associated with the development or suppression of various types of cancer, including PCa. This review summarizes the recent findings on m6A regulation and its functions and mechanisms in cells, focusing on the various functional proteins operating within m6A in PCa cells. Moreover, the potential clinical value of exploiting m6A modification as an early diagnostic marker in PCa diagnosis and therapeutics was discussed. m6A may also be used as an indicator to evaluate treatment outcome and prognosis.
Collapse
Affiliation(s)
- Hongyuan Wan
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Yanyan Feng
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Junjie Wu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Lijie Zhu
- Department of Urology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Yuanyuan Mi
- Department of Urology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| |
Collapse
|
6
|
Bi JH, Jiang YH, Ye SJ, Wu MR, Yi Y, Wang HX, Wang LM. Investigation of the inhibition effect of 1,2,3,4,6-pentagalloyl-β-D-glucose on gastric cancer cells based on a network pharmacology approach and experimental validation. Front Oncol 2022; 12:934958. [PMID: 35992839 PMCID: PMC9383036 DOI: 10.3389/fonc.2022.934958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGastric cancer (GC) is ranked as the third leading cause of cancer-related mortality worldwide. 1,2,3,4,6-Pentagalloyl-β-D-glucose (β-PGG) has various pharmacological activities and has been shown to suppress cancer development. However, the mechanism by which β-PGG inhibits gastric cancer has not been elucidated.ObjectiveThis study explored the potential targets and mechanism of β-PGG in GC using the network pharmacology approach combined with in-vitro experiments.MethodsThe PharmMapper software was used to predict the potential targets of β-PGG, and GC-related genes were identified on the GeneCards database. PPI analysis of common genes was performed using the STRING database. The potential regulatory mechanism of β-PGG in GC was explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The binding ability of key genes and target proteins was verified by molecular docking. The effects of β-PGG on genes and proteins were evaluated using the CCK-8 assay, cell cycle analysis, apoptosis assay, real-time fluorescence quantification polymerase chain reaction (qRT-PCR), and Western blotting.ResultsEight hub genes involved in cell cycle progression and apoptosis were identified. Cancer-related signaling pathways were identified using the Cytoscape tool. Some of those genes were significantly enriched in the p53 signaling pathway. The CCK-8 assay showed that β-PGG inhibited the proliferation of GC cells. Cell cycle and apoptosis experiments revealed that β-PGG induced cell cycle arrest and apoptosis of gastric cancer cells. qRT-PCR and Western blot analysis showed that β-PGG inhibited β-PGG cells by modulating the p53 signaling pathway.ConclusionIn the present study, the targets and mechanism of β-PGG in gastric cancer were explored. The results indicate that β-PGG can be used to develop treatments for GC.
Collapse
Affiliation(s)
- Jing-hui Bi
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Yu-han Jiang
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Shi-jie Ye
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Min-rui Wu
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Yang Yi
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hong-xun Wang
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Li-mei Wang
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
- *Correspondence: Li-mei Wang,
| |
Collapse
|
7
|
Fan CW, Tang J, Jiang JC, Zhou MM, Li MS, Wang HS. Pentagalloylglucose suppresses the growth and migration of human nasopharyngeal cancer cells via the GSK3β/β-catenin pathway in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154192. [PMID: 35636179 DOI: 10.1016/j.phymed.2022.154192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a type of malignant squamous cell tumour originating from the nasopharynx epithelium. Pentagalloylglucose (PGG) is a natural polyphenolic compound that exerts anticancer effects in many types of tumours. However, the role and underlying mechanism of PGG in NPC cells have not been fully defined. PURPOSE This study aimed to investigate the anticancer activity of PGG as well as the potential mechanism in NPC cells. METHODS The effects of PGG on the proliferation, apoptosis and cell cycle distribution of CNE1 and CNE2 cells were assessed by MTT and flow cytometry assays. Cell migration was evaluated using wound healing and transwell assays. The expression of microtubule-associated protein 1 light chain 3 beta (LC3B) was observed by immunofluorescence staining. Western blotting was used to explore the levels of related proteins and signalling pathway components. Furthermore, the effects of PGG on NPC cell growth were analysed in a xenograft mouse model in vivo using cisplatin as a positive control. RESULTS PGG dose-dependently inhibited the proliferation of CNE1 and CNE2 cells. PGG regulated the cell cycle by altering p53, cyclin D1, CDK2, and cyclin E1 protein levels. PGG induced apoptosis and autophagy in NPC cells and elevated the Bax/Bcl-2 ratio and the protein levels of LC3B. Moreover, PGG decreased NPC cell migration by increasing E-cadherin and decreasing N-cadherin, vimentin and CD44 protein levels. Mechanistically, PGG treatment downregulated p-mTOR and β-catenin expression but upregulated p-p38 MAPK and p-GSK3β expression. In addition, PGG significantly inhibited NPC cell tumour growth and lung metastasis in vivo. CONCLUSION PGG may suppress cell proliferation, induce apoptosis and autophagy, and decrease the metastatic capacity of NPC cells through the p38 MAPK/mTOR and Wnt/β-catenin pathways. The present study provides evidence for PGG as a potential therapy for NPC.
Collapse
Affiliation(s)
- Cai-Wen Fan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China; Research Center for Science, Guilin Medical University, Guilin 541199, China
| | - Juan Tang
- Department of Pathology, the Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China
| | - Jing-Chen Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Mei-Mei Zhou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Mei-Shan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| | - Heng-Shan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
8
|
Li J, Yu W, Ge J, Zhang J, Wang Y, Wang P, Shi G. Targeting eIF3f Suppresses the Growth of Prostate Cancer Cells by Inhibiting Akt Signaling. Onco Targets Ther 2020; 13:3739-3750. [PMID: 32440143 PMCID: PMC7210466 DOI: 10.2147/ott.s244345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/10/2020] [Indexed: 01/14/2023] Open
Abstract
Background Eukaryotic initiation factor 3 (eIF3) is the largest translation initiation factor, and oncogenic roles have been discovered for its subunits, including the f subunit (ie, eIF3f), in various human cancers. However, the roles of eIF3f in the development and progression of prostate cancer (PCa) have not been reported. Materials and Methods We performed in silico analysis to screen the expression of eIF3 subunits. Relevant shRNAs were used to knock down eIF3 subunits in 22Rv1 cells and cell proliferation was analyzed. eIF3f expression in PCa specimens was confirmed by immunohistochemistry. eIF3f knockdown was established to evaluate the effects of eIF3f on cell proliferation in vitro and in vivo. RNA‐seq, bioinformatics analysis and Western blotting were applied to explore the molecular details underlying the biological function of eIF3f in PCa cells. shRNA-resistant eIF3f and myristoylated-Akt were used to rescue the effects of eIF3f disturbance on PCa cells. Results Functional analyses confirmed that eIF3f is essential for PCa proliferation. Notably, the expression of eIF3f was found to be elevated in human PCa tissues as well as in PCa cell lines. eIF3f silencing significantly suppressed the growth of PCa cells, both in vitro and in vivo. eIF3f expression was positively correlated with Akt signaling activity in RNA-seq profiles and published prostate cohorts. Knockdown of eIF3f markedly reduced the levels of phosphorylated Akt in PCa cells. Exogenous expression of shRNA-resistant eIF3f in eIF3f knockdown cells restored Akt phosphorylation levels and cell growth. Importantly, rescue experiments revealed that ectopic expression of myristoylated-Akt partially alleviated the suppressive effects of eIF3f disturbance with respect to the growth of PCa cells. Conclusion These results suggested that eIF3f has an oncogenic role in PCa, mediated at least partially through the regulation of Akt signaling, and that eIF3f represents a potential target for the inhibition of PCa growth and progression.
Collapse
Affiliation(s)
- Junhong Li
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Wandong Yu
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Jianchao Ge
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Jun Zhang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Yang Wang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Pengyu Wang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| | - Guowei Shi
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, People's Republic of China
| |
Collapse
|
9
|
Kantapan J, Paksee S, Chawapun P, Sangthong P, Dechsupa N. Pentagalloyl Glucose- and Ethyl Gallate-Rich Extract from Maprang Seeds Induce Apoptosis in MCF-7 Breast Cancer Cells through Mitochondria-Mediated Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:5686029. [PMID: 32382295 PMCID: PMC7193289 DOI: 10.1155/2020/5686029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/26/2020] [Accepted: 03/25/2020] [Indexed: 01/26/2023]
Abstract
Bouea macrophylla Griffith, locally known as maprang, has important economic value as a Thai fruit tree. The maprang seed extract (MPSE) has been shown to exhibit antibacterial and anticancer activities. However, the bioactive constituents in MPSE and the molecular mechanisms underlying these anticancer activities remain poorly understood. This study aims to identify the active compounds in MPSE and to investigate the mechanisms involved in MPSE-induced apoptosis in MCF-7 treated cancer cells. The cytotoxic effect was determined by MTT assay. The apoptosis induction of MPSE was evaluated in terms of ROS production, mitochondrial membrane potential depolarization, and apoptosis-related gene expression. The compounds identified by HPLC and LC/MS analysis were pentagalloyl glucose, ethyl gallate, and gallic acid. MPSE treatment decreased cell proliferation in MCF-7 cells, and MPSE was postulated to induce G2/M phase cell cycle arrest. MPSE was found to promote intracellular ROS production in MCF-7 treated cells and to also influence the depolarization of mitochondrial membrane potential. In addition, MPSE treatment can lead to increase in the Bax/Bcl-2 gene expression ratio, suggesting that MPSE-induced apoptosis is mitochondria-dependent pathway. Our results suggest that natural products obtained from maprang seeds have the potential to target the apoptosis pathway in breast cancer treatments.
Collapse
Affiliation(s)
- Jiraporn Kantapan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siwaphon Paksee
- Department of Radiological Technology, Kanchanabhishek Institute of Medical and Public Health Technology, Nonthaburi 11150, Thailand
| | - Pornthip Chawapun
- Interdisciplinary Program in Biotechnology, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center on Chemistry for Development of Health Promoting Products from Northern Resources, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nathupakorn Dechsupa
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
10
|
Won YS, Seo KI. Sanggenol L Induces Apoptosis and Cell Cycle Arrest via Activation of p53 and Suppression of PI3K/Akt/mTOR Signaling in Human Prostate Cancer Cells. Nutrients 2020; 12:nu12020488. [PMID: 32075054 PMCID: PMC7071324 DOI: 10.3390/nu12020488] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/22/2020] [Accepted: 02/10/2020] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is the most common cancer in Western countries. Recently, Asian countries are being affected by Western habits, which have had an important role in the rapid increase in cancer incidence. Sanggenol L (San L) is a natural flavonoid present in the root barks of Morus alba, which induces anti-cancer activities in ovarian cancer cells. However, the molecular and cellular mechanisms of the effects of sanggenol L on human prostate cancer cells have not been elucidated. In this study, we investigated whether sanggenol L exerts anti-cancer activity in human prostate cancer cells via apoptosis and cell cycle arrest. Sanggenol L induced caspase-dependent apoptosis (up-regulation of PARP and Bax or down-regulation of procaspase-3, -8, -9, Bid, and Bcl-2), induction of caspase-independent apoptosis (up-regulation of AIF and Endo G on cytosol), suppression of cell cycle (down-regulation of CDK1/2, CDK4, CDK6, cyclin D1, cyclin E, cyclin A, and cyclin B1 or up-regulation of p53 and p21), and inhibition of PI3K/Akt/mTOR signaling (down-regulation of PI3K, p-Akt, and p-mTOR) in prostate cancer cells. These results suggest the induction of apoptosis via suppression of PI3K/Akt/mTOR signaling and cell cycle arrest via activation of p53 in response to sanggenol L in prostate cancer cells.
Collapse
|
11
|
Xiao W, Zhang W, Huang H, Xie Y, Zhang Y, Guo X, Jin C, Liao X, Yao S, Chen G, Song X. Cancer Targeted Gene Therapy for Inhibition of Melanoma Lung Metastasis with eIF3i shRNA Loaded Liposomes. Mol Pharm 2019; 17:229-238. [PMID: 31765158 DOI: 10.1021/acs.molpharmaceut.9b00943] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Eukaryotic translation initiation factors 3i (eIF3i) is a proto-oncogene that is overexpressed in various tumors, reducing its expression by eIF3i shRNA is a promising strategy to inhibit tumor growth or metastasis. Tumor cell is the target of eIF3i shRNA so that tumor-site accumulation could be important for fulfilling its therapeutic effect. Thus, the iRGD modified liposome (R-LP) was rationally synthesized to enhance the antitumor effect by active targeted delivery of eIF3i shRNA to B16F10 melanoma cells. R-LP encapsulating eIF3i shRNA gene (R-LP/sheIF3i) were prepared by a film dispersion method. The transfection experiment proves that R-LP could effectively transfect B16F10 cells. R-LP/sheIF3i notably restrained the migration, invasion, and adhesion of melanoma cells in vitro. In a mouse model of lung metastasis, R-LP/sheIF3i administered by intravenous injection suppressed pulmonary metastasis of melanoma by dramatically downregulated eIF3i expression and subsequently inhibiting tumor neovascularization and tumor cells proliferation in vivo. Our results provide a basis for tumor cells targeting strategies to reduce the expression of eIF3i by RNAi in the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Wen Xiao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Weiyi Zhang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Hai Huang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Yafei Xie
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Yi Zhang
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xia Guo
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Chaohui Jin
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xuelian Liao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Shaohua Yao
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Guo Chen
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| | - Xiangrong Song
- State Key Laboratory of Biotherapy/Geriatrics and Cancer Center, West China Hospital and Collaborative Innovation Center for Biotherapy , Sichuan University , Chengdu , 610041 , China
| |
Collapse
|
12
|
Kaplan A, Kutlu HM, Ciftci GA. Fe 3O 4 Nanopowders: Genomic and Apoptotic Evaluations on A549 Lung Adenocarcinoma Cell Line. Nutr Cancer 2019; 72:708-721. [PMID: 31335223 DOI: 10.1080/01635581.2019.1643031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The magnetite nanoparticles are progressively used in a wide range of biological applications. In the present study, we purposed to show apoptosis-inducing ability of Fe3O4 nanopowders on A549 cells. In addition, the toxic effects of Fe3O4 nanopowders were researched on L929 cells. The cytotoxicity of Fe3O4 nanopowders were evaluated on A549 and L929 cells by MTT assay and inhibited cell proliferation by time and dose-dependent manner on A549 cells but was not toxic on L929 cells. According to these findings, IC30 value of Fe3O4 nanopowders was determined as 5 µM. The early and late apoptotic cells were detected by Annexin V-FITC/PI assay using IC30 concentration of Fe3O4 nanopowders. Furthermore, The IC30 value of Fe3O4 nanopowders was not effective in the activation of caspase-3 but was effective on loss of mitochondrial membrane potential. The apoptotic index of A549 cells was investigated and found out to increase by IC30 value of Fe3O4 nanopowders using TUNEL, BrdU, Bcl-2 immunocytochemical assays. The upregulated and downregulated genes were profiled and the presence of some apoptotic genes was determined with administration of IC30 value of Fe3O4 nanopowders by microarray assay. This work suggests that Fe3O4 nanopowders could be a good candidate for therapy of lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Ayse Kaplan
- Department of Biology, Faculty of Science, Eskisehir Technical University, Eskisehir, Turkey
| | - Hatice Mehtap Kutlu
- Department of Biology, Faculty of Science, Eskisehir Technical University, Eskisehir, Turkey
| | - Gulsen Akalin Ciftci
- Faculty of Pharmacy, Department of Biochemistry, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
13
|
Small molecule inhibition of matrix metalloproteinases as a potential therapeutic for metastatic activity in squamous cell carcinoma. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s41548-019-00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Xiang Q, Tang J, Luo Q, Xue J, Tao Y, Jiang H, Tian J, Fan C. In vitro study of anti-ER positive breast cancer effect and mechanism of 1,2,3,4-6-pentyl-O-galloyl-beta-d-glucose (PGG). Biomed Pharmacother 2019; 111:813-820. [DOI: 10.1016/j.biopha.2018.12.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 01/16/2023] Open
|
15
|
Zhang R, Yu R, Xu Q, Li X, Luo J, Jiang B, Wang L, Guo S, Wu N, Shi D. Discovery and evaluation of the hybrid of bromophenol and saccharide as potent and selective protein tyrosine phosphatase 1B inhibitors. Eur J Med Chem 2017; 134:24-33. [PMID: 28395151 DOI: 10.1016/j.ejmech.2017.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/21/2017] [Accepted: 04/02/2017] [Indexed: 11/28/2022]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a key negative regulator of insulin signaling pathway. Inhibition of PTP1B is expected to improve insulin action. Appropriate selectivity and permeability are the gold standard for excellent PTP1B inhibitors. In this work, molecular hybridization-based screening identified a selective competitive PTP1B inhibitor. Compound 10a has IC50 values of 199 nM against PTP1B, and shows 32-fold selectivity for PTP1B over the closely related phosphatase TCPTP. Molecule docking and molecular dynamics studies reveal the reason of selectivity for PTP1B over TCPTP. Moreover, the cell permeability and cellular activity of compound 10a are demonstrated respectively.
Collapse
Affiliation(s)
- Renshuai Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qi Xu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiangqian Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jiao Luo
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bo Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Lijun Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shuju Guo
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Li JZ, Wang ZL, Xu WH, Li Q, Gao L, Wang ZM. MicroRNA-495 Regulates Migration and Invasion in Prostate Cancer Cells Via Targeting Akt and mTOR Signaling. Cancer Invest 2016; 34:181-8. [PMID: 27031291 DOI: 10.3109/07357907.2016.1156690] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abnormal microRNA (miR) expressions were implicated in prostate cancer progression. We identified a novel miR-495, which was downregulated in prostate cancer, but not normal prostate cell lines. MiR-495 directly targeted the 3'-UTR of Akt and mTOR mRNAs. Expression of miR-495 in prostate cancer cells significantly downregulated Akt and mTOR, which further inhibited cancer cell proliferation, migration, and invasion in vitro. Function of miR-495 in vivo was examined in mouse xenograft model and was found to significantly inhibit the growth of tumors, mediated by repressing Akt and mTOR. Our report supported miR-495 as a novel tumor suppressor microRNA in prostate cancer.
Collapse
Affiliation(s)
- Jian-Zhang Li
- a Department of Urology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Zhen-Long Wang
- b Department of Urology , The Second Affiliated Hospital of Xi'an Jiaotong University , Xi'an , China
| | - Wan-Hai Xu
- a Department of Urology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Qing Li
- a Department of Urology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Lin Gao
- a Department of Urology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Zi-Ming Wang
- b Department of Urology , The Second Affiliated Hospital of Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|
17
|
Meng Y, Lin ZM, Ge N, Zhang DL, Huang J, Kong F. Ursolic Acid Induces Apoptosis of Prostate Cancer Cells via the PI3K/Akt/mTOR Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1471-86. [PMID: 26503559 DOI: 10.1142/s0192415x15500834] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ursolic acid (UA), a pentacyclic triterpenoid, is known to exert antitumor activity in breast, lung, liver and colon cancers. Nonetheless, the underlying mechanism of ursolic acid in prostate cancer cells still remains unclear. In the present study, we report the chemotherapeutic effects of ursolic acid as assessed using in vitro and in vivo models. Treatment of human prostate cancer cells (LNCaP and PC-3) with UA inhibited the proliferation and induced apoptosis in both cell lines as characterized by the increased Annexin V-binding. The induction of apoptosis by UA was associated with a decrease in the levels of Bcl-2, Bcl-xl, survivin, and activated caspase-3. Treatment with UA also inhibited the expression of phosphatidylinositol-3-kinase (PI3K), phosphorylation of Akt and mTOR signaling proteins. Further, administration of UA significantly inhibited the growth of LNCaP prostate tumor xenografts in athymic nude mice, which was associated with inhibition of cell proliferation, induction of apoptosis of tumor cells and decreased expression of PI3K downstream factors, such as p-Akt and p-mTOR in tumor xenograft tissues. Our study demonstrates that UA not only inhibits cell growth but also induces apoptosis through modulation of the PI3K/Akt/mTOR pathway in human prostate cancer cells. We suggest that UA may be a new chemotherapeutic candidate against prostate cancer.
Collapse
Affiliation(s)
- Yan Meng
- * Department of Urology, 247 Beiyuan Road, Jinan 250033, Shandong, P.R. China
| | - Zhao-Min Lin
- † Central Research Laboratory, Second Hospital of Shandong University, 247 Beiyuan Road, Jinan 250033, Shandong, P.R. China
| | - Nan Ge
- * Department of Urology, 247 Beiyuan Road, Jinan 250033, Shandong, P.R. China
| | - Deng-Lu Zhang
- * Department of Urology, 247 Beiyuan Road, Jinan 250033, Shandong, P.R. China
| | - Jie Huang
- ‡ Shandong Medical Imaging Research Institute, 324 Jingwu Road, Jinan 250021, Shandong, P.R. China
| | - Feng Kong
- † Central Research Laboratory, Second Hospital of Shandong University, 247 Beiyuan Road, Jinan 250033, Shandong, P.R. China
| |
Collapse
|