1
|
Zheng Y, Ma Y, Xiong Q, Zhu K, Weng N, Zhu Q. The role of artificial intelligence in the development of anticancer therapeutics from natural polyphenols: Current advances and future prospects. Pharmacol Res 2024; 208:107381. [PMID: 39218422 DOI: 10.1016/j.phrs.2024.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Natural polyphenols, abundant in the human diet, are derived from a wide variety of sources. Numerous preclinical studies have demonstrated their significant anticancer properties against various malignancies, making them valuable resources for drug development. However, traditional experimental methods for developing anticancer therapies from natural polyphenols are time-consuming and labor-intensive. Recently, artificial intelligence has shown promising advancements in drug discovery. Integrating AI technologies into the development process for natural polyphenols can substantially reduce development time and enhance efficiency. In this study, we review the crucial roles of natural polyphenols in anticancer treatment and explore the potential of AI technologies to aid in drug development. Specifically, we discuss the application of AI in key stages such as drug structure prediction, virtual drug screening, prediction of biological activity, and drug-target protein interaction, highlighting the potential to revolutionize the development of natural polyphenol-based anticancer therapies.
Collapse
Affiliation(s)
- Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Yifei Ma
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Kai Zhu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Ningna Weng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2
|
Abid F, Saleem M, Leghari T, Rafi I, Maqbool T, Fatima F, Arshad AM, Khurshid S, Naz S, Hadi F, Tahir M, Akhtar S, Yasir S, Mobashar A, Ashraf M. Evaluation of in vitro anticancer potential of pharmacological ethanolic plant extracts Acacia modesta and Opuntia monocantha against liver cancer cells. BRAZ J BIOL 2024; 84:e252526. [DOI: 10.1590/1519-6984.252526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/27/2021] [Indexed: 12/30/2022] Open
Abstract
Abstract Acacia modesta (AM) and Opuntia monocantha (OM) are distributed in Pakistan, Afghanistan and India. Both of these plants have different pharmacological properties. This study was designed to evaluate anticancer potential of Acacia modesta (AM) and Opuntia monocantha (OM). Liver cancer cell line HepG2 was used for assessment of anticancer activity. For the evaluation of anti-proliferative effects, cell viability and cell death in all groups of cells were evaluated via MTT, crystal violet and trypan blue assays. For the evaluation of apoptosis ELISA of p53 performed. Furthermore, LDH assay to find out the ability of malignant cells to metabolize pyruvate to lactate and antioxidant enzymes activity (GSH, CAT and SOD) at the end HPLC was performed to find active compound of AM and OM. Cytotoxicity (MTT), Viability assays (trypan blue, crystal viability, MUSE analysis) showed more dead, less live cells in plant treated groups with increase of concentration. Scratch assay for the anti-migratory effect of these plants showed treated groups have not ability to heal scratch/wound. ELISA of p53 for cellular apoptosis showed more release of p53 in treated groups. Antioxidant assay via glutathione (GSH), superoxide dismutase (SOD), catalase (CAT) showed less anti-oxidative potential in treated cancer groups. LDH assay showed more lactate dehydrogenase release in treated groups compared with untreated. HPLC analysis showed the presence of phytochemicals such as steroids, alkaloids, phenols, flavonoids, saponins, tannins, anthraquinone and amino acids in AM and OM plant extracts. Based on all these findings, it can be concluded that ethanolic extracts of Acacia modesta and Opuntia monocantha have promising anti-cancer potential.
Collapse
Affiliation(s)
- F. Abid
- Government College University Faisalabad, Pakistan; The University of Lahore, Pakistan
| | - M. Saleem
- Government College University Faisalabad, Pakistan; University of the Punjab, Pakistan
| | | | - I. Rafi
- University of Lahore, Pakistan
| | | | | | | | | | - S. Naz
- University of Lahore, Pakistan
| | - F. Hadi
- University of Lahore, Pakistan
| | | | - S. Akhtar
- University of Lahore, Pakistan; University of Bradford, United Kingdom
| | | | | | | |
Collapse
|
3
|
Si W, Zhang Y, Li X, Du Y, Xu Q. Understanding the Functional Activity of Polyphenols Using Omics-Based Approaches. Nutrients 2021; 13:nu13113953. [PMID: 34836207 PMCID: PMC8625961 DOI: 10.3390/nu13113953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023] Open
Abstract
Plant polyphenols are the main category of natural active substances, and are distributed widely in vegetables, fruits, and plant-based processed foods. Polyphenols have a beneficial performance in preventing diseases and maintaining body health. However, its action mechanism has not been well understood. Foodomics is a novel method to sequence and widely used in nutrition, combining genomics, proteomics, transcriptomics, microbiome, and metabolomics. Based on multi-omics technologies, foodomics provides abundant data to study functional activities of polyphenols. In this paper, physiological functions of various polyphenols based on foodomics and microbiome was discussed, especially the anti-inflammatory and anti-tumor activities and gut microbe regulation. In conclusion, omics (including microbiomics) is a useful approach to explore the bioactive activities of polyphenols in the nutrition and health of human and animals.
Collapse
Affiliation(s)
- Wenjin Si
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.S.); (X.L.); (Y.D.)
- Shennongjia Science & Technology Innovation Center, Huazhong Agricultural University, Wuhan 430070, China
| | - Yangdong Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
| | - Xiang Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.S.); (X.L.); (Y.D.)
- Shennongjia Science & Technology Innovation Center, Huazhong Agricultural University, Wuhan 430070, China
| | - Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.S.); (X.L.); (Y.D.)
- Shennongjia Science & Technology Innovation Center, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.S.); (X.L.); (Y.D.)
- Shennongjia Science & Technology Innovation Center, Huazhong Agricultural University, Wuhan 430070, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
- Correspondence:
| |
Collapse
|
4
|
Wei T, Lambert PF. Role of IQGAP1 in Carcinogenesis. Cancers (Basel) 2021; 13:3940. [PMID: 34439095 PMCID: PMC8391515 DOI: 10.3390/cancers13163940] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/31/2022] Open
Abstract
Scaffolding proteins can play important roles in cell signaling transduction. IQ motif-containing GTPase-activating protein 1 (IQGAP1) influences many cellular activities by scaffolding multiple key signaling pathways, including ones involved in carcinogenesis. Two decades of studies provide evidence that IQGAP1 plays an essential role in promoting cancer development. IQGAP1 is overexpressed in many types of cancer, and its overexpression in cancer is associated with lower survival of the cancer patient. Here, we provide a comprehensive review of the literature regarding the oncogenic roles of IQGAP1. We start by describing the major cancer-related signaling pathways scaffolded by IQGAP1 and their associated cellular activities. We then describe clinical and molecular evidence for the contribution of IQGAP1 in different types of cancers. In the end, we review recent evidence implicating IQGAP1 in tumor-related immune responses. Given the critical role of IQGAP1 in carcinoma development, anti-tumor therapies targeting IQGAP1 or its associated signaling pathways could be beneficial for patients with many types of cancer.
Collapse
Affiliation(s)
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| |
Collapse
|
5
|
Zhao Q, Peng C, Zheng C, He XH, Huang W, Han B. Recent Advances in Characterizing Natural Products that Regulate Autophagy. Anticancer Agents Med Chem 2020; 19:2177-2196. [PMID: 31749434 DOI: 10.2174/1871520619666191015104458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/16/2018] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Autophagy, an intricate response to nutrient deprivation, pathogen infection, Endoplasmic Reticulum (ER)-stress and drugs, is crucial for the homeostatic maintenance in living cells. This highly regulated, multistep process has been involved in several diseases including cardiovascular and neurodegenerative diseases, especially in cancer. It can function as either a promoter or a suppressor in cancer, which underlines the potential utility as a therapeutic target. In recent years, increasing evidence has suggested that many natural products could modulate autophagy through diverse signaling pathways, either inducing or inhibiting. In this review, we briefly introduce autophagy and systematically describe several classes of natural products that implicated autophagy modulation. These compounds are of great interest for their potential activity against many types of cancer, such as ovarian, breast, cervical, pancreatic, and so on, hoping to provide valuable information for the development of cancer treatments based on autophagy.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Xiang-Hong He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China.,The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, United States
| |
Collapse
|
6
|
Brisdelli F, Di Francesco L, Giorgi A, Lizzi AR, Luzi C, Mignogna G, Bozzi A, Schininà ME. Proteomic Analysis of Quercetin-Treated K562 Cells. Int J Mol Sci 2019; 21:ijms21010032. [PMID: 31861640 PMCID: PMC6981597 DOI: 10.3390/ijms21010032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Among natural products under investigation for their additive potential in cancer prevention and treatment, the flavonoid quercetin has received attention for its effects on the cell cycle arrest and apoptosis. In the past, we addressed this issue in K562 cells, a cellular model of the human chronic myeloid leukemia. Here, we applied stable isotope labeling by amino acids in cell culture (SILAC) proteomics with the aim to increase knowledge on the regulative and metabolic pathways modulated by quercetin in these cells. After 24 h of quercetin treatment, we observed that apoptosis was not completely established, thus we selected this time range to capture quantitative data. As a result, we were able to achieve a robust identification of 1703 proteins, and to measure fold changes between quercetin-treated and untreated cells for 1206 proteins. Through a bioinformatics functional analysis on a subset of 112 proteins, we propose that the apoptotic phenotype of K562 cells entails a significant modulation of the translational machinery, RNA metabolism, antioxidant defense systems, and enzymes involved in lipid metabolism. Finally, we selected eight differentially expressed proteins, validated their modulated expression in quercetin-treated K562 cells, and discussed their possible role in flavonoid cytotoxicity. This quantitative profiling, performed for the first time on this type of tumor cells upon treatment with a flavonoid, will contribute to revealing the molecular basis of the multiplicity of the effects selectively exerted by quercetin on K562 cells.
Collapse
Affiliation(s)
- Fabrizia Brisdelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.B.); (A.R.L.); (C.L.); (A.B.)
| | - Laura Di Francesco
- Department of Biochemical Sciences, Sapienza, University of Rome, 00185 Rome, Italy; (L.D.F.); (A.G.); (G.M.)
| | - Alessandra Giorgi
- Department of Biochemical Sciences, Sapienza, University of Rome, 00185 Rome, Italy; (L.D.F.); (A.G.); (G.M.)
| | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.B.); (A.R.L.); (C.L.); (A.B.)
| | - Carla Luzi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.B.); (A.R.L.); (C.L.); (A.B.)
| | - Giuseppina Mignogna
- Department of Biochemical Sciences, Sapienza, University of Rome, 00185 Rome, Italy; (L.D.F.); (A.G.); (G.M.)
| | - Argante Bozzi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.B.); (A.R.L.); (C.L.); (A.B.)
| | - M. Eugenia Schininà
- Department of Biochemical Sciences, Sapienza, University of Rome, 00185 Rome, Italy; (L.D.F.); (A.G.); (G.M.)
- Correspondence:
| |
Collapse
|
7
|
Kerimi A, Williamson G. Differential Impact of Flavonoids on Redox Modulation, Bioenergetics, and Cell Signaling in Normal and Tumor Cells: A Comprehensive Review. Antioxid Redox Signal 2018; 29:1633-1659. [PMID: 28826224 PMCID: PMC6207159 DOI: 10.1089/ars.2017.7086] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Flavonoids can interact with multiple molecular targets to elicit their cellular effects, leading to changes in signal transduction, gene expression, and/or metabolism, which can, subsequently, affect the entire cell and organism. Immortalized cell lines, derived from tumors, are routinely employed as a surrogate for mechanistic studies, with the results extrapolated to tissues in vivo. Recent Advances: We review the activities of selected flavonoids on cultured tumor cells derived from various tissues in comparison to corresponding primary cells or tissues in vivo, mainly using quercetin and flavanols (epicatechin and (-)-epigallocatechin gallate) as exemplars. Several studies have indicated that flavonoids could retard cancer progression in vivo in animal models as well as in tumor cell models. CRITICAL ISSUES Extrapolation from in vitro and animal models to humans is not straightforward given both the extensive conjugation and complex microbiota-dependent metabolism of flavonoids after consumption, as well as the heterogeneous metabolism of different tumors. FUTURE DIRECTIONS Comparison of data from studies on primary cells or in vivo are essential not only to validate results obtained from cultured cell models, but also to highlight whether any differences may be further exploited in the clinical setting for chemoprevention. Tumor cell models can provide a useful mechanistic tool to study the effects of flavonoids, provided that the limitations of each model are understood and taken into account in interpretation of the data.
Collapse
Affiliation(s)
- Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds , Leeds, United Kingdom
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
8
|
Chen Y, Wang X, Cao C, Wang X, Liang S, Peng C, Fu L, He G. Inhibition of HSP90 sensitizes a novel Raf/ERK dual inhibitor CY-9d in triple-negative breast cancer cells. Oncotarget 2017; 8:104193-104205. [PMID: 29262632 PMCID: PMC5732798 DOI: 10.18632/oncotarget.22119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 09/22/2017] [Indexed: 02/05/2023] Open
Abstract
Raf and extracellular signal-regulated kinases (ERK) are both important therapeutic targets in the mitogen-activated protein kinase (MAPK) pathway, and play crucial roles in the apoptosis resistance of breast cancer cells. In the present study, cytotoxic and apoptosis-inducing activities of the Raf/ERK dual inhibitor CY-9d were found to be restricted in triple negative breast cancer (TNBC) cells compared with ER/PR-positive cells. Based on the analysis of differentially expressed proteins using a quantitative proteomic iTRAQ method and bioinformatics analysis, HSP90 was found to identify as a potential mediator between Raf and ERK in TNBC cells. Western blotting and RNA interference suggested that down-regulated IQGAP1 can attenuate the routine Raf/MEK/ERK cascade and recruit HSP90 as a bypass pathway. Simultaneous treatment with the HSP90 inhibitor and CY-9d at sub-therapeutic doses was found to produce synergistic therapeutic and apoptosis-inducing effects in TNBC cells. Moreover, CY-9d was also found to suppress breast cancer growth, inhibit the activation of Raf/ERK, and induce mitochondrial apoptosis in vivo without remarkable toxicity. These results support the combination of HSP90 and Raf/ERK inhibitors as a potential target therapeutic strategy with enhanced tumor growth suppression, downstream pathway blockade, and greater induction of apoptosis.
Collapse
Affiliation(s)
- Yujuan Chen
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xiaoyun Wang
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Chuan Cao
- State Key Laboratory Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaodong Wang
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Leilei Fu
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Gu He
- State Key Laboratory of Biotherapy and Department of Breast Surgery, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| |
Collapse
|
9
|
Proteomic evaluation of mouse adipose tissue and liver following hydroxytyrosol supplementation. Food Chem Toxicol 2017; 107:329-338. [DOI: 10.1016/j.fct.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 12/16/2022]
|
10
|
Liu J, He Y, Zhang D, Cai Y, Zhang C, Zhang P, Zhu H, Xu N, Liang S. In vitro anticancer effects of two novel phenanthroindolizidine alkaloid compounds on human colon and liver cancer cells. Mol Med Rep 2017; 16:2595-2603. [PMID: 28677760 PMCID: PMC5548052 DOI: 10.3892/mmr.2017.6879] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/07/2017] [Indexed: 02/05/2023] Open
Abstract
Malignant cancer is one of the most serious diseases threatening the health of human beings. Natural plant alkaloids exhibit multiple biological functions, including inhibition of cell proliferation, and may have potential anticancer activity. However, most natural alkaloids may not be suitable for human therapies owing to instability, poor dissolubility and potential side effects. To improve their anticancer activity and drug effect, the present study aimed to develop new alkaloid derivatives, the phenanthroindolizidine alkaloid compounds, and evaluated their potential antitumor effects on human cancer cells in vitro. Among the several newly synthesized analogues of phenanthroindolizidine alkaloids (PAs), the compounds YS306 and YS206 exhibited an increased growth inhibition activity on HepG2 liver cancer cells and on HCT116 and HT29 colon cancer cells, with half‑maximal inhibitory concentrations in the micromolar range. YS206 and YS306 (5 µg/ml) both significantly induced cell cycle arrest at the G2/M phase and notably decreased cell distribution at the G0/G1 and S phase. In addition, these two molecules significantly inhibited cancer cell migration, as analyzed by the wound‑healing and Transwell assays. However, neither YS306 nor YS206 exhibited observable effects on apoptosis. Therefore, chemical structure modifications of natural PAs based on anticancer activity assessments may be feasible in the development of new cancer chemotherapeutic agents.
Collapse
Affiliation(s)
- Jingjing Liu
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Yu He
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Dan Zhang
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Ying Cai
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Chenggang Zhang
- Department of Chemistry and Materials, Sichuan Normal University, Chengdu, Sichuan 610066, P.R. China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 100034, P.R. China
| | - Ningzhi Xu
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Shufang Liang
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
11
|
Understanding the Effectiveness of Natural Compound Mixtures in Cancer through Their Molecular Mode of Action. Int J Mol Sci 2017; 18:ijms18030656. [PMID: 28304343 PMCID: PMC5372668 DOI: 10.3390/ijms18030656] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 12/19/2022] Open
Abstract
Many approaches to cancer management are often ineffective due to adverse reactions, drug resistance, or inadequate target specificity of single anti-cancer agents. In contrast, a combinatorial approach with the application of two or more anti-cancer agents at their respective effective dosages can achieve a synergistic effect that boosts cytotoxicity to cancer cells. In cancer, aberrant apoptotic pathways allow cells that should be killed to survive with genetic abnormalities, leading to cancer progression. Mutations in apoptotic mechanism arising during the treatment of cancer through cancer progression can consequently lead to chemoresistance. Natural compound mixtures that are believed to have multiple specific targets with minimal acceptable side-effects are now of interest to many researchers due to their cytotoxic and chemosensitizing activities. Synergistic interactions within a drug mixture enhance the search for potential molecular targets in cancer cells. Nonetheless, biased/flawed scientific evidence from natural products can suggest false positive therapeutic benefits during drug screening. In this review, we have taken these factors into consideration when discussing the evidence for these compounds and their synergistic therapeutic benefits in cancer. While there is limited evidence for clinical efficacy for these mixtures, in vitro data suggest that these preparations merit further investigation, both in vitro and in vivo.
Collapse
|
12
|
Zhou J, Fang L, Liao J, Li L, Yao W, Xiong Z, Zhou X. Investigation of the anti-cancer effect of quercetin on HepG2 cells in vivo. PLoS One 2017; 12:e0172838. [PMID: 28264020 PMCID: PMC5338765 DOI: 10.1371/journal.pone.0172838] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/10/2017] [Indexed: 12/18/2022] Open
Abstract
Quercetin, a natural polyphenolic flavonoid compound, can inhibit the growth of several malignant cancers. However, the mechanism still remains unclear. Our previous findings have suggested that quercetin can significantly inhibit HepG2 cell proliferation and induce cell apoptosis in vitro. It can also affect cell cycle distribution and significantly decrease cyclin D1 expression. In this study, we investigated the anti-cancer effect of quercetin on HepG2 tumor-bearing nude mice and its effect on cyclin D1 expression in the tumor tissue. First, the nude murine tumor model was established by subcutaneous inoculation of HepG2 cells, then quercetin was administered intraperitoneally, and the mice injected with saline solution were used as controls. The daily behavior of the tumor-bearing mice was observed and differences in tumor growth and survival rate were monitored. The expression of cyclin D1 in isolated tumor sections was evaluated by immunohistochemistry. We found that HepG2 tumor became palpable in the mice one-week post-inoculation. Tumors in the control group grew rapidly and the daily behavior of the mice changed significantly, including listlessness, poor feeding and ataxia. The mice in quercetin-treated group showed delayed tumor growth, no significant changes in daily behavior, and the survival rate was significantly improved. Finally, we observed increased tumor necrosis and a lighter cyclin D1 staining with reduced staining areas. Our findings thus suggest that quercetin can significantly inhibit HepG2 cell proliferation, and this effect may be achieved through the regulation of cyclin D1 expression.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
- * E-mail:
| | - Li Fang
- Department of Gastroenterology, the First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Jiaxu Liao
- Department of Radiology, the Sixth Hospital, Chengdu, Sichuan, P.R. China
| | - Lin Li
- Department of Nuclear Medicine, the Second Hospital, Chengdu, Sichuan, P.R. China
| | - Wenxiu Yao
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Zhujuan Xiong
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Xiang Zhou
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| |
Collapse
|
13
|
Zoheir KM, Abd-Rabou AA, Harisa GI, Kumar A, Ahmad SF, Ansari MA, Abd-Allah AR. IQGAP1 gene silencing induces apoptosis and decreases the invasive capacity of human hepatocellular carcinoma cells. Tumour Biol 2016; 37:13927-13939. [PMID: 27488117 DOI: 10.1007/s13277-016-5283-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/15/2016] [Indexed: 12/24/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) belong to a conserved family, and they are involved in various intracellular processes. IQGAP1 is expressed in all cells, while IQGAP2 and IQGAP3 are mainly expressed in hepatic cells. IQGAP1 has been suggested to be an oncogene, while IQGAP2 is considered a tumor-suppressor gene. However, the relationship between RAS family genes and IQGAP genes remains unclear. We recently demonstrated this interaction in a chemically induced mouse liver cancer. In this study, IQGAP1 expression was partially silenced in human hepatocellular carcinoma (HepG2) cells. We investigated the impact of IQGAP1 silencing on the interactions of IQGAP and RAS with several apoptotic proteins, including caspase-3 (CASP3), BCL2-associated X protein (BAX), and B-cell leukemia/lymphoma 2 (BCL2). Additionally, we investigated the effects of the interactions of these genes on cell viability, proliferation, apoptosis, and invasive capacity. IQGAP1 siRNA-treated HepG2 cells showed lower invasive capacity than the control cells, and this reduction was time- and vector concentration-dependent. In addition, IQGAP1 silencing resulted in significantly lower IQGAP1 level and subsequently higher IQGAP2 and IQGAP3 expression in HepG2 cells than in the control. Flow cytometry analyses indicated that the silencing of IQGAP1 can induce early and late apoptosis in HepG2 cells. Additionally, IQGAP2, IQGAP3, CASP3, and BAX were upregulated whereas IQGAP1 and BCL2 were downregulated in the siRNA-treated cells. Furthermore, we observed that the mRNA levels of HRAS, KRAS, NRAS, and MRAS decreased upon IQGAP1 silencing. These findings indicate that IQGAP1 potentially regulates the expression of IQGAP and RAS gene families and demonstrate its regulatory role in the apoptotic network. Taken together, our findings suggest that IQGAP1 silencing plays crucial roles in the apoptosis of HepG2 cells and lowers their proliferative and invasive capacities.
Collapse
Affiliation(s)
- Khairy Ma Zoheir
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia. .,Cell Biology Department, National Research Centre, Cairo, 12622, Egypt.
| | - Ahmed A Abd-Rabou
- Hormones Department, Medical Research Division, National Research Centre, Cairo, 12622, Egypt
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, PO Box 11451, Riyadh, Saudi Arabia
| | - Ashok Kumar
- Vitiligo Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh Fayaz Ahmad
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq Ahmad Ansari
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Adel R Abd-Allah
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
14
|
Zhou J, Li LU, Fang LI, Xie H, Yao W, Zhou X, Xiong Z, Wang LI, Li Z, Luo F. Quercetin reduces cyclin D1 activity and induces G1 phase arrest in HepG2 cells. Oncol Lett 2016; 12:516-522. [PMID: 27347174 PMCID: PMC4906932 DOI: 10.3892/ol.2016.4639] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/29/2016] [Indexed: 02/05/2023] Open
Abstract
Quercetin is able to inhibit proliferation of malignant tumor cells; however, the exact mechanism involved in this biological process remains unclear. The current study utilized a quantitative proteomic analysis to explore the antitumor mechanisms of quercetin. The leucine of HepG2 cells treated with quercetin was labeled as d3 by stable isotope labeling by amino acids in cell culture (SILAC). The isotope peaks of control HepG2 cells were compared with the d3-labeled HepG2 cells by mass spectrometry (MS) to identify significantly altered proteins. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analyses were subsequently employed to verify the results of the MS analysis. A flow cytometry assay was designed to observe the influence of various quercetin treatment concentrations on the cell cycle distribution of HepG2 cells. The results indicated that quercetin is able to substantially inhibit proliferation of HepG2 cells and induce an obvious morphological alteration of cells. According to the MS results, the 70 credibly-changed proteins that were identified may play important roles in multiple cellular processes, including protein synthesis, signaling, cytoskeletal processes and metabolism. Among these functional proteins, the expression of cyclin D1 (CCND1) was found to be significantly decreased. RT-PCR and western blot analyses verified the SILAC-MS results of decreased CCND1 expression. In summary, flow cytometry revealed that quercetin is able to induce G1 phase arrest in HepG2 cells. Based on the aforementioned observations, it is suggested that quercetin exerts antitumor activity in HepG2 cells through multiple pathways, including interfering with CCND1 gene expression to disrupt the cell cycle and proliferation of HepG2 cells. In the future, we aim to explore this effect in vivo.
Collapse
Affiliation(s)
- Jin Zhou
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China; Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - L U Li
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - L I Fang
- Department of Gastroenterology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Hua Xie
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Wenxiu Yao
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Xiang Zhou
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Zhujuan Xiong
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - L I Wang
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhixi Li
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Luo
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
15
|
Pi J, Li B, Tu L, Zhu H, Jin H, Yang F, Bai H, Cai H, Cai J. Investigation of quercetin-induced HepG2 cell apoptosis-associated cellular biophysical alterations by atomic force microscopy. SCANNING 2016; 38:100-12. [PMID: 26179807 DOI: 10.1002/sca.21245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/28/2015] [Accepted: 06/04/2015] [Indexed: 05/15/2023]
Abstract
Quercetin, a wildly distributed bioflavonoid, has been proved to possess excellent antitumor activity on hepatocellular carcinoma (HCC). In the present study, the biophysical properties of HepG2 cells were qualitatively and quantitatively determined using high resolution atomic force microscopy (AFM) to understand the anticancer effects of quercetin on HCC cells at nanoscale. The results showed that quercetin could induce severe apoptosis in HepG2 cells through arrest of cell cycle and disruption of mitochondria membrane potential. Additionally, the nuclei and F-actin structures of HepG2 cells were destroyed by quercetin treatment as well. AFM morphological data showed some typical apoptotic characterization of HepG2 cells with increased particle size and roughness in the ultrastructure of cell surface upon quercetin treatment. As an important biophysical property of cells, the membrane stiffness of HepG2 cells was further quantified by AFM force measurements, which indicated that HepG2 cells became much stiffer after quercetin treatment. These results collectively suggest that quercetin can be served as a potential therapeutic agent for HCC, which not only extends our understanding of the anticancer effects of quercetin against HCC cells into nanoscale, but also highlights the applications of AFM for the investigation of anticancer drugs.
Collapse
Affiliation(s)
- Jiang Pi
- State Key Laboratory of Quality Research in Chinese, Macau University of Science and Technology, Macau, China
| | - Baole Li
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Lvying Tu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Haiyan Zhu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Hua Jin
- State Key Laboratory of Quality Research in Chinese, Macau University of Science and Technology, Macau, China
| | - Fen Yang
- State Key Laboratory of Quality Research in Chinese, Macau University of Science and Technology, Macau, China
| | - Haihua Bai
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Huaihong Cai
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese, Macau University of Science and Technology, Macau, China
- Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Proteomic Analysis of Anticancer TCMs Targeted at Mitochondria. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:539260. [PMID: 26568766 PMCID: PMC4629060 DOI: 10.1155/2015/539260] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/30/2015] [Indexed: 12/16/2022]
Abstract
Traditional Chinese medicine (TCM) is a rich resource of anticancer drugs. Increasing bioactive natural compounds extracted from TCMs are known to exert significant antitumor effects, but the action mechanisms of TCMs are far from clear. Proteomics, a powerful platform to comprehensively profile drug-regulated proteins, has been widely applied to the mechanistic investigation of TCMs and the identification of drug targets. In this paper, we discuss several bioactive TCM products including terpenoids, flavonoids, and glycosides that were extensively investigated by proteomics to illustrate their antitumor mechanisms in various cancers. Interestingly, many of these natural compounds isolated from TCMs mostly exert their tumor-suppressing functions by specifically targeting mitochondria in cancer cells. These TCM components induce the loss of mitochondrial membrane potential, the release of cytochrome c, and the accumulation of ROS, initiating apoptosis cascade signaling. Proteomics provides systematic views that help to understand the molecular mechanisms of the TCM in tumor cells; it bears the inherent limitations in uncovering the drug-protein interactions, however. Subcellular fractionation may be coupled with proteomics to capture and identify target proteins in mitochondria-enriched lysates. Furthermore, translating mRNA analysis, a new technology profiling the drug-regulated genes in translatome level, may be integrated into the systematic investigation, revealing global information valuable for understanding the action mechanism of TCMs.
Collapse
|
17
|
Bi Y, Shen C, Li C, Liu Y, Gao D, Shi C, Peng F, Liu Z, Zhao B, Zheng Z, Wang X, Hou X, Liu H, Wu J, Zou H, Wang K, Zhong C, Zhang J, Shi C, Zhao S. Inhibition of autophagy induced by quercetin at a late stage enhances cytotoxic effects on glioma cells. Tumour Biol 2015; 37:3549-60. [PMID: 26454746 DOI: 10.1007/s13277-015-4125-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/20/2015] [Indexed: 01/08/2023] Open
Abstract
Glioma is the most common primary brain tumor in the central nervous system (CNS) with high morbidity and mortality in adults. Although standardized comprehensive therapy has been adapted, the prognosis of glioma patients is still frustrating and thus novel therapeutic strategies are urgently in need. Quercetin (Quer), an important flavonoid compound found in many herbs, is shown to be effective in some tumor models including glioma. Recently, it is reported that adequate regulation of autophagy can strengthen cytotoxic effect of anticancer drugs. However, it is not yet fully clear how we should modulate autophagy to achieve a satisfactory therapeutic effect. 3-Methyladenine (3-MA) and Beclin1 short hairpin RNA (shRNA) were used to inhibit the early stage of autophage while chloroquine (CQ) to inhibit the late stage. MTT assay was implemented to determine cell viability. Transmission electron microscopy, western blot, and immunohistochemistry were adopted to evaluate autophagy. Western blot, flow cytometry, and immunohistochemistry were used to detect apoptosis. C6 glioma xenograft models were established to assess the therapeutic effect (the body weight change, the median survival time, and tumor volume) in vivo. Quercetin can inhibit cell viability and induce autophagy of U87 and U251 glioma cells in a dose-dependent manner. Inhibition of early-stage autophagy by 3-MA or shRNA against Beclin1 attenuated the quercetin-induced cytotoxicity. In contrast, suppression of autophagy at a late stage by CQ enhanced the anti-glioma efficiency of quercetin. Therapeutic effect of quercetin for malignant glioma can be strengthened by inhibition of autophagy at a late stage, not initial stage, which may provide a novel opportunity for glioma therapy.
Collapse
Affiliation(s)
- Yunke Bi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Chen Shen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Chenguang Li
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Yaohua Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Dandan Gao
- Department of Endocrinology, General Hospital of HeiLongJiang Agricultural Reclamation Department, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Chen Shi
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fei Peng
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Zhendong Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Boxian Zhao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Zhixing Zheng
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Xiaoxiong Wang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Xu Hou
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Huailei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Jianing Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Huichao Zou
- Department of Pain Medicine, Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Kaikai Wang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Chen Zhong
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Jiakang Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Changbin Shi
- Section of Neurosurgery, Department of Surgery, The University of Chicago Medical Center and Pritzker School of Medicine, Chicago, IL, USA
| | - Shiguang Zhao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China. .,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.
| |
Collapse
|
18
|
Abstract
Food-derived flavonoid quercetin, widely distributed in onions, apples, and tea, is able to inhibit growth of various cancer cells indicating that this compound can be considered as a good candidate for anticancer therapy. Although the exact mechanism of this action is not thoroughly understood, behaving as antioxidant and/or prooxidant as well as modulating different intracellular signalling cascades may all play a certain role. Such inhibitory activity of quercetin has been shown to depend first of all on cell lines and cancer types; however, no comprehensive site-specific analysis of this effect has been published. In this review article, cytotoxicity constants of quercetin measured in various human malignant cell lines of different origin were compiled from literature and a clear cancer selective action was demonstrated. The most sensitive malignant sites for quercetin revealed to be cancers of blood, brain, lung, uterine, and salivary gland as well as melanoma whereas cytotoxic activity was higher in more aggressive cells compared to the slowly growing cells showing that the most harmful cells for the organism are probably targeted. More research is needed to overcome the issues of poor water solubility and relatively low bioavailability of quercetin as the major obstacles limiting its clinical use.
Collapse
|
19
|
Chen JY, Hu RY, Chou HC. Quercetin-induced cardioprotection against doxorubicin cytotoxicity. J Biomed Sci 2013; 20:95. [PMID: 24359494 PMCID: PMC3898810 DOI: 10.1186/1423-0127-20-95] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/11/2013] [Indexed: 11/19/2022] Open
Abstract
Background Cancer has continually been the leading cause of death worldwide for decades. Thus, scientists have actively devoted themselves to studying cancer therapeutics. Doxorubicin is an efficient drug used in cancer therapy, but also produces reactive oxygen species (ROS) that induce severe cytotoxicity against heart cells. Quercetin, a plant-derived flavonoid, has been proven to contain potent antioxidant and anti-inflammatory properties. Thus, this in vitro study investigated whether quercetin can decrease doxorubicin-induced cytotoxicity and promote cell repair systems in cardiomyocyte H9C2 cells. Results Proteomic analysis and a cell biology assay were performed to investigate the quercetin-induced responses. Our data demonstrated that quercetin treatment protects the cardiomyocytes in a doxorubicin-induced heart damage model. Quercetin significantly facilitated cell survival by inhibiting cell apoptosis and maintaining cell morphology by rearranging the cytoskeleton. Additionally, 2D-DIGE combined with MALDI-TOF MS analysis indicated that quercetin might stimulate cardiomyocytes to repair damage after treating doxorubicin by modulating metabolic activation, protein folding and cytoskeleton rearrangement. Conclusion Based on a review of the literature, this study is the first to report detailed protective mechanisms for the action of quercetin against doxorubicin-induced cardiomyocyte toxicity based on in-depth cell biology and proteomic analysis.
Collapse
Affiliation(s)
| | | | - Hsiu-Chuan Chou
- Department of Applied Science, National Hsinchu University of Education, Hsinchu, Taiwan.
| |
Collapse
|
20
|
Quantitative secretome analysis reveals the interactions between epithelia and tumor cells by in vitro modulating colon cancer microenvironment. J Proteomics 2013; 89:51-70. [DOI: 10.1016/j.jprot.2013.05.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/22/2013] [Accepted: 05/20/2013] [Indexed: 11/18/2022]
|
21
|
Li H, Li G, Zhao X, Wu Y, Ma W, Liu Y, Gong F, Liang S. Complementary serum proteomic analysis of autoimmune hepatitis in mice and patients. J Transl Med 2013; 11:146. [PMID: 23763817 PMCID: PMC3702393 DOI: 10.1186/1479-5876-11-146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022] Open
Abstract
Background Autoimmune hepatitis (AIH) is a chronic liver disease caused by inflammation of the liver. The etiology of AIH remains elusive, and there are no reliable serum biomarkers. Methods In order to identify candidate biomarkers, 2-DE analysis of serum proteins was performed using a mouse model of AIH induced by treatment with concanavalin A (ConA). To enrich samples for low abundance molecules a commercial albumin removal reagent was used. In an independent analysis, candidate biomarkers were identified in AIH patient’s serum by a targeted iTRAQ (isobaric tags for relative and absolute quantification) identification. Candidates were validated in independent cohorts of ConA treated mice and AIH patients by ELISA (enzyme-linked immuno sorbent assay). Results Nine proteins were differentially expressed in AIH mice treated with con-A. Two of these, the third component of complement (C3) and alpha-2-macroglobulin (A2M) were also up-regulated in AIH patient’s sera by a targeted iTRAQ identification. In separate validation studies, serum C3 and A2M levels were increased in mice with ConA treatment after 20-40 h and in 34 AIH patients in a subgroup analysis, females with AIH aged 20–50 years old displayed the largest increases in serum A2M level. Biological network analysis implements the complement cascade and protease inhibitors in the pathogenesis of AIH. Conclusion The serum proteins C3 and A2M are increased both in a mouse model and in patients with AIH by both 2-DE and iTRAQ methods. This integrated serum proteomics investigation should be applicable for translational researchers to study other medical conditions.
Collapse
|
22
|
Zhang L, Li X, Zhang L, Wang B, Zhang T, Ye J. Chlamydophila (Chlamydia) pneumoniae infection promotes vascular smooth muscle cell adhesion and migration through IQ domain GTPase-activating protein 1. Microb Pathog 2012; 53:207-13. [PMID: 22835851 DOI: 10.1016/j.micpath.2012.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/14/2012] [Accepted: 07/17/2012] [Indexed: 01/16/2023]
Abstract
The mechanisms for Chlamydophila (Chlamydia) pneumoniae (C. pneumoniae) infection-induced atherosclerosis are still unclear. Cell adhesion has important roles in vascular smooth muscle cell (VSMC) migration required in the development of atherosclerosis. However, it is still unknown whether IQ domain GTPase-activating protein 1 (IQGAP1) plays pivotal roles in C. pneumoniae infection-induced the adhesion and migration of rat primary VSMCs. Accordingly, in this study, we demonstrated that rat primary VSMC adhesion (P < 0.001) and migration (P < 0.01) measured by cell adhesion assay and Transwell assay, respectively, were significantly enhanced after C. pneumoniae infection. Reverse transcription-polymerase chain reaction analysis revealed that the mRNA expression levels of IQGAP1 in the infected rat primary VSMCs were found to increase gradually to reach a peak and then decrease gradually to a level similar to the control. We further showed that the increases in rat primary VSMC adhesion to Matrigel (P < 0.001) and migration (P < 0.01) caused by C. pneumoniae infection were markedly inhibited after IQGAP1 knockdown by a pool of four short hairpin RNAs. Taken together, our results suggest that C. pneumoniae infection may promote the adhesion and migration of VSMCs possibly by upregulating the IQGAP1 expression.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | | | | | | | | | | |
Collapse
|
23
|
Romagnolo DF, Milner JA. Opportunities and challenges for nutritional proteomics in cancer prevention. J Nutr 2012; 142:1360S-9S. [PMID: 22649262 DOI: 10.3945/jn.111.151803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Knowledge gaps persist about the efficacy of cancer prevention strategies based on dietary food components. Adaptations to nutrient supply are executed through tuning of multiple protein networks that include transcription factors, histones, modifying enzymes, translation factors, membrane and nuclear receptors, and secreted proteins. However, the simultaneous quantitative and qualitative measurement of all proteins that regulate cancer processes is not practical using traditional protein methodologies. Proteomics offers an attractive opportunity to fill this knowledge gap and unravel the effects of dietary components on protein networks that impinge on cancer. The articles presented in this supplement are from talks proffered in the "Nutrition Proteomics and Cancer Prevention" session at the American Institute for Cancer Research Annual Research Conference on Food, Nutrition, Physical Activity and Cancer held in Washington, DC on October 21 and 22, 2010. Recent advances in MS technologies suggest that studies in nutrition and cancer prevention may benefit from the adoption of proteomic tools to elucidate the impact on biological processes that govern the transition from normal to malignant phenotype; to identify protein changes that determine both positive and negative responses to food components; to assess how protein networks mediate dose-, time-, and tissue-dependent responses to food components; and, finally, for predicting responders and nonresponders. However, both the limited accessibility to proteomic technologies and research funding appear to be hampering the routine adoption of proteomic tools in nutrition and cancer prevention research.
Collapse
Affiliation(s)
- Donato F Romagnolo
- Department of Nutritional Sciences and The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ, USA.
| | | |
Collapse
|
24
|
Yang JD, Sun Z, Hu C, Lai J, Dove R, Nakamura I, Lee JS, Thorgeirsson SS, Kang KJ, Chu IS, Roberts LR. Sulfatase 1 and sulfatase 2 in hepatocellular carcinoma: associated signaling pathways, tumor phenotypes, and survival. Genes Chromosomes Cancer 2011; 50:122-135. [PMID: 21104785 PMCID: PMC3253341 DOI: 10.1002/gcc.20838] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The heparin-degrading endosulfatases sulfatase 1 (SULF1) and sulfatase 2 (SULF2) have opposing effects in hepatocarcinogenesis despite structural similarity. Using mRNA expression arrays, we analyzed the correlations of SULF expression with signaling networks in human hepatocellular carcinomas (HCCs) and the associations of SULF expression with tumor phenotype and patient survival. Data from two mRNA microarray analyses of 139 and 36 HCCs and adjacent tissues were used as training and validation sets. Partek and Metacore software were used to identify SULF correlated genes and their associated signaling pathways. Associations between SULF expression, the hepatoblast subtype of HCC, and survival were examined. Both SULF1 and 2 had strong positive correlations with periostin, IQGAP1, TGFB1, and vimentin and inverse correlations with HNF4A and IQGAP2. Genes correlated with both SULFs were highly associated with the cell adhesion, cytoskeletal remodeling, blood coagulation, TGFB, and Wnt/β-catenin and epithelial mesenchymal transition signaling pathways. Genes uniquely correlated with SULF2 were more associated with neoplastic processes than genes uniquely correlated with SULF1. High SULF expression was associated with the hepatoblast subtype of HCC. There was a bimodal effect of SULF1 expression on prognosis, with patients in the lowest or highest tertile having a worse prognosis than those in the middle tertile. SULFs have complex effects on HCC signaling and patient survival. There are functionally similar associations with cell adhesion, ECM remodeling, TGFB, and WNT pathways, but also unique associations of SULF1 and SULF2. The roles and targeting of the SULFs in cancer require further investigation.
Collapse
Affiliation(s)
- Ju Dong Yang
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Zhifu Sun
- Department of Biomedical Statistics and Informatics, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Chunling Hu
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Jinping Lai
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Rebecca Dove
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Ikuo Nakamura
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | | | - Koo Jeong Kang
- Surgery, Keimyung University School of Medicine, Daegu, Korea
| | - In-Sun Chu
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Lewis R. Roberts
- Miles and Shirley Fiterman Center for Digestive Diseases, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| |
Collapse
|
25
|
Liang S, Fu A, Zhang Q, Tang M, Zhou J, Wei Y, Chen L. Honokiol inhibits HepG2 migration via down-regulation of IQGAP1 expression discovered by a quantitative pharmaceutical proteomic analysis. Proteomics 2010; 10:1474-83. [PMID: 20127691 DOI: 10.1002/pmic.200900649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Honokiol (HNK), a natural small molecular product, inhibited proliferation of HepG2 cells and exhibited anti-tumor activity in nude mice. In this article, we applied a novel sensitive stable isotope labeling with amino acids in cell culture-based quantitative proteomic method and a model of nude mice to investigate the correlation between HNK and the hotspot migration molecule Ras GTPase-activating-like protein (IQGAP1). The quantitative proteomic analysis showed that IQGAP1 was 0.53-fold down-regulated under 10 microg/mL HNK exposure for 24 h on HepG2 cells. Migration ability of HepG2 cells under HNK treatment was correlated with its expression level of IQGAP1. In addition, the biochemical validation on HepG2 cells and the tumor xenograft model further demonstrated that HNK decreased the expression level of IQGAP1 and its upstream proteins Cdc42/Rac1. These data supported that HNK can modulate cell adhesion and cell migration by acting on Cdc42/Rac1 signaling via IQGAP1 interactions with its upstream Cdc42/Rac1 proteins, which is a new molecular mechanism of HNK to exert its anti-tumor activity.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, West China Medical School, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
26
|
Baik JY, Lee GM. A DIGE approach for the assessment of differential expression of the CHO proteome under sodium butyrate addition: Effect of Bcl-xLoverexpression. Biotechnol Bioeng 2010; 105:358-67. [DOI: 10.1002/bit.22534] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
White CD, Brown MD, Sacks DB. IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis. FEBS Lett 2009; 583:1817-24. [PMID: 19433088 DOI: 10.1016/j.febslet.2009.05.007] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 04/28/2009] [Accepted: 05/02/2009] [Indexed: 12/13/2022]
Abstract
The IQGAP family comprises three proteins in humans. The best characterized is IQGAP1, which participates in protein-protein interactions and integrates diverse signaling pathways. IQGAP2 and IQGAP3 harbor all the domains identified in IQGAP1, but their biological roles are poorly defined. Proteins that bind IQGAP1 include Cdc42 and Rac1, E-cadherin, beta-catenin, calmodulin and components of the mitogen-activated protein kinase pathway, all of which are involved in cancer. Here, we summarize the biological functions of IQGAPs that may contribute to neoplasia. Additionally, we review published data which implicate IQGAPs in cancer and tumorigenesis. The cumulative evidence suggests IQGAP1 is an oncogene while IQGAP2 may be a tumor suppressor.
Collapse
Affiliation(s)
- Colin D White
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|