1
|
Costa Cerqueira M, Silva A, Martins Sousa S, Pinto-Ribeiro F, Baltazar F, Afonso J, Freitas Costa M. Chromene-based compounds as drug candidates for renal and bladder cancer therapy - A systematic review. Bioorg Chem 2024; 153:107865. [PMID: 39393199 DOI: 10.1016/j.bioorg.2024.107865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
Renal (RC) and bladder cancers (BC) are common urological malignancies prevalent in the male population. Incidence and mortality rates are expected to increase in the near future. Drug toxicity and development of drug resistance in both diseases are major obstacles to achieve successful treatments. Chromenes are heterocyclic compounds constituted by a benzene ring fused to a pyran nucleus. Natural and synthetic chromene-based compounds have proven to be promising anticancer agents. Additionally, re-sensitization of cancer cells to classical treatments has also been demonstrated. Thus, the aim of this systematic review is to assess the potential of chromene-based compounds in the treatment of RC and BC. Study collection was performed in six different databases, to compile existing information on preclinical (in vitro and in vivo) and clinical studies developed to date. Overall, multiple chromene-based compounds showed potent anticancer effects, affecting several biological features such as reduction in cell viability, proliferation, migration and invasion in vitro, and induction of cell cycle arrest and cell death. Tumor volume and weight were generally decreased in vivo upon chromene-based treatment. Modest results have been obtained in two clinical trials, with reports of a partial response and two objective responses in RC patients. Thus, the chromene family can be considered an attractive chemical scaffold, harboring promising drug candidates for RC and BC therapeutics.
Collapse
Affiliation(s)
- Mónica Costa Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Ana Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sofia Martins Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Julieta Afonso
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Marta Freitas Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal.
| |
Collapse
|
2
|
Singh D, Shukla G. The multifaceted anticancer potential of luteolin: involvement of NF-κB, AMPK/mTOR, PI3K/Akt, MAPK, and Wnt/β-catenin pathways. Inflammopharmacology 2024:10.1007/s10787-024-01596-8. [PMID: 39543054 DOI: 10.1007/s10787-024-01596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Cancer is the predominant and major cause of fatality worldwide, based on the different types of cancer. There is a limitation in the current treatment. So we need better therapeutic agents that counteract the progression and development of malignant tumours. Plant-derived products are closely related and useful for human health. Luteolin is a polyphenolic flavonoid bioactive molecule that is present in various herbs, vegetables, fruits, and flowers and exhibits chemoprotective and pharmacological activity against different malignancies. To offer innovative approaches for the management of various cancers, we present a comprehensive analysis of the latest discoveries on luteolin. The aim is to inspire novel concepts for the development of advanced pharmaceuticals targeting cancer and search specifically targeted reviews and research articles published from January 1999 to January 2024 that investigated the application of luteolin in various cancer management. A thorough literature search utilizing the keywords "luteolin" "Signalling Pathway" "cancer" and nanoparticles was performed in the databases of Google Scholar, Web of Science, SCOPUS, UGC care list and PubMed. Through the compilation of existing research, we have discovered that luteolin possesses several therapeutic actions against various cancer via a signaling pathway involving the of NF-κB regulation, AMPK/mTOR, toll-like receptor, Nrf-2, PI3K/Akt MAPK and Wnt/β-catenin and their underlying mechanism of action has been well understood. This review intended to completely integrate crucial information on natural sources, biosynthesis, pharmacokinetics, signaling pathways, chemoprotective and therapeutic properties against various cancers, and nanoformulation of luteolin.
Collapse
Affiliation(s)
- Deepika Singh
- Faculty of Health Sciences, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India.
| | - Gaurav Shukla
- Faculty of Health Sciences, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| |
Collapse
|
3
|
Ou YC, Yu TM, Li JR, Wu CC, Wang JD, Liao SL, Chen WY, Kuan YH, Chen CJ. Runx2 silencing sensitized human renal cell carcinoma cells to ABT-737 apoptosis. Arch Biochem Biophys 2024; 761:110173. [PMID: 39369835 DOI: 10.1016/j.abb.2024.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
The prognostic value of Runt-related transcription factor 2 (Runx2) and its involvement in cell growth and motility have been reported in patients diagnosed with renal cell carcinoma (RCC). Since Runx2 may have the potential to be a target for the purpose of antitumor intervention, there is an urgent need to gain insight into its oncogenic properties. Using human 786-O, Caki-1 and ACHN RCC cells as models, the silencing of cellular Runx2 expression brought about a reduction in cyclin D1 and β-catenin expression, cell growth and migration without any significant cell death. Runx2-silenced cells turned into apoptosis vulnerable in the presence of ABT-737, a BH3 mimetic Bcl-2 inhibitor. Data from biochemical and molecular studies have revealed a positive correlation between Runx2 expression and Akt phosphorylation, Mcl-1 expression, and fibronectin expression. Results of genetic silencing studies have indicated the potential involvement of Mcl-1 and fibronectin in the decision of RCC cell ABT-737 resistance and sensitivity. The regulatory roles of the PI3K/Akt axis in the expression of Mcl-1 and fibronectin were suggested by means of the results taken from experiments involving pharmacological study of the PI3K/Akt. Since overexpression and prognostic roles of Runx2, activated Akt, Mcl-1, fibronectin, cyclin D1, and β-catenin have been revealed in RCC, it is important to explore the precise mechanisms underlying Runx2 oncogenic effects. Although the linking details between Runx2 and PI3K/Akt have yet to be identified, our findings suggest that Mcl-1 and fibronectin are downstream effectors of Runx2 via a regulatory axis of the PI3K/Akt and their promotion of cell growth, migration, and ABT-737 resistance in RCC cells.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung City, 433, Taiwan.
| | - Tung-Min Yu
- Division of Nephrology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan; Department of Financial Engineering, Providence University, Taichung City, 433, Taiwan; Department of Data Science and Big Data Analytics, Providence University, Taichung City, 433, Taiwan.
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan.
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung City, 402, Taiwan.
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, 407, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, 404, Taiwan.
| |
Collapse
|
4
|
Govindasamy B, Muthu M, Gopal J, Chun S. A review on the impact of TRAIL on cancer signaling and targeting via phytochemicals for possible cancer therapy. Int J Biol Macromol 2023; 253:127162. [PMID: 37788732 DOI: 10.1016/j.ijbiomac.2023.127162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 09/11/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023]
Abstract
Anticancer therapies have been the continual pursuit of this age. Cancer has been ravaging all across the globe breathing not just threats but demonstrating them. Remedies for cancer have been frantically sought after. Few have worked out, yet till date, the available cancer therapies have not delivered a holistic solution. In a world where the search for therapies is levitating towards natural remedies, solutions based on phytochemicals are highly prospective attractions. A lot has been achieved with inputs from plant resources, providing numerous natural remedies. In the current review, we intensely survey the progress achieved in the treatment of cancer through phytochemicals-based programmed cell death of cancer cells. More specifically, we have further reviewed and discussed the role of phytochemicals in activating apoptosis via Tumor Necrosis Factor-Alpha-Related Apoptosis-Inducing Ligand (TRAIL), which is a cell protein that can attach to certain molecules in cancer cells, killing cancer cells. The objective of this review is to enlist the various phytochemicals that are available for specifically contributing towards triggering the TRAIL cell protein-mediated cancer therapy and to point out the research gaps that require future research motivation. This is the first review of this kind in this research direction.
Collapse
Affiliation(s)
- Balasubramani Govindasamy
- Department of Product Development, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, India
| | - Manikandan Muthu
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, India
| | - Judy Gopal
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, India
| | - Sechul Chun
- Department of Bioresources and Food Science, Institute of Natural Science and Agriculture, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
5
|
Luiz-Ferreira A, Pacifico T, Cruz ÁC, Laudisi F, Monteleone G, Stolfi C. TRAIL-Sensitizing Effects of Flavonoids in Cancer. Int J Mol Sci 2023; 24:16596. [PMID: 38068921 PMCID: PMC10706592 DOI: 10.3390/ijms242316596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
Collapse
Affiliation(s)
- Anderson Luiz-Ferreira
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Álefe Cardoso Cruz
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| |
Collapse
|
6
|
Bajalia EM, Azzouz FB, Chism DA, Giansiracusa DM, Wong CG, Plaskett KN, Bishayee A. Phytochemicals for the Prevention and Treatment of Renal Cell Carcinoma: Preclinical and Clinical Evidence and Molecular Mechanisms. Cancers (Basel) 2022; 14:3278. [PMID: 35805049 PMCID: PMC9265746 DOI: 10.3390/cancers14133278] [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] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Renal cell carcinoma (RCC) is associated with about 90% of renal malignancies, and its incidence is increasing globally. Plant-derived compounds have gained significant attention in the scientific community for their preventative and therapeutic effects on cancer. To evaluate the anticancer potential of phytocompounds for RCC, we compiled a comprehensive and systematic review of the available literature. Our work was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses criteria. The literature search was performed using scholarly databases such as PubMed, Scopus, and ScienceDirect and keywords such as renal cell carcinoma, phytochemicals, cancer, tumor, proliferation, apoptosis, prevention, treatment, in vitro, in vivo, and clinical studies. Based on in vitro results, various phytochemicals, such as phenolics, terpenoids, alkaloids, and sulfur-containing compounds, suppressed cell viability, proliferation and growth, showed cytotoxic activity, inhibited invasion and migration, and enhanced the efficacy of chemotherapeutic drugs in RCC. In various animal tumor models, phytochemicals suppressed renal tumor growth, reduced tumor size, and hindered angiogenesis and metastasis. The relevant antineoplastic mechanisms involved upregulation of caspases, reduction in cyclin activity, induction of cell cycle arrest and apoptosis via modulation of a plethora of cell signaling pathways. Clinical studies demonstrated a reduced risk for the development of kidney cancer and enhancement of the efficacy of chemotherapeutic drugs. Both preclinical and clinical studies displayed significant promise of utilizing phytochemicals for the prevention and treatment of RCC. Further research, confirming the mechanisms and regulatory pathways, along with randomized controlled trials, are needed to establish the use of phytochemicals in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (E.M.B.); (F.B.A.); (D.A.C.); (D.M.G.); (C.G.W.); (K.N.P.)
| |
Collapse
|
7
|
Han S, Lin F, Qi Y, Liu C, Zhou L, Xia Y, Chen K, Xing J, Liu Z, Yu W, Zhang Y, Zhou X, Rao T, Cheng F. HO-1 Contributes to Luteolin-Triggered Ferroptosis in Clear Cell Renal Cell Carcinoma via Increasing the Labile Iron Pool and Promoting Lipid Peroxidation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3846217. [PMID: 35656025 PMCID: PMC9153929 DOI: 10.1155/2022/3846217] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/26/2022] [Indexed: 12/16/2022]
Abstract
Ferroptosis, a novel form of regulated cell death characterized by disrupted iron metabolism and the accumulation of lipid peroxides, has exhibited enormous potential in the therapy of cancer particularly clear cell renal cell carcinoma (ccRCC). Luteolin (Lut), a natural flavonoid widely existing in various fruits and vegetables, has been proven to exert potent anticancer activity in vitro and in vivo. However, previous studies on the anticancer mechanism of Lut have been shown in apoptosis but not ferroptosis. In the present study, we identified that Lut substantially inhibited the survival of ccRCC in vitro and in vivo, and this phenomenon was accompanied by excessively increased intracellular Fe2+ and abnormal depletion of GSH. In addition, Lut induced the imbalance of mitochondrial membrane potential, classical morphological alterations of mitochondrial ferroptosis, generation of ROS, and occurrence of lipid peroxidation in an iron-dependent manner in ccRCC cells. However, these alterations induced by Lut could be reversed to some extent by the iron ion chelator deferiprone or the ferroptosis inhibitor ferrostatin-1, indicating that ccRCC cells treated with Lut underwent ferroptosis. Mechanistically, molecular docking further established that Lut probably promoted the heme degradation and accumulation of labile iron pool (LIP) by excessively upregulating the HO-1 expression, which led to the Fenton reaction, GSH depletion, and lipid peroxidation in ccRCC, whereas blocking this signaling pathway evidently rescued the Lut-induced cell death of ccRCC by inhibiting ferroptosis. Altogether, the current study shows that the natural compound monomer Lut exerted anticancer efficacy by excessively upregulating HO-1 expression and activating LIP to trigger ferroptosis in ccRCC and could be a promising and potent drug candidate for ccRCC treatment.
Collapse
Affiliation(s)
- Shangting Han
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yucheng Qi
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Cong Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Linxiang Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kang Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ji Xing
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zilin Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yunlong Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
8
|
Molecular Mechanism of Gleditsiae Spina for the Treatment of High-Grade Serous Ovarian Cancer Based on Network Pharmacology and Pharmacological Experiments. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5988310. [PMID: 35299895 PMCID: PMC8923798 DOI: 10.1155/2022/5988310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/13/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022]
Abstract
Background Gleditsiae Spina, widely used in traditional Chinese medicine, has a good curative effect on malignant tumors such as ovarian cancer, but the mechanism is not clear. So, we aimed to analyze the pharmacological mechanism of Gleditsiae Spina in the treatment of high-grade serous ovarian cancer (HGSC) based on network pharmacology and biological experiments. Methods The main active ingredients of Gleditsiae Spina were identified by high performance liquid chromatography (HPLC) and mass spectrometry (MS), and the active ingredients were performed by ADME screening. The component targets of Gleditsiae Spina were screened using the PharmMapper platform, and differentially expressed genes in normal and HGSC tissues were identified through the GEO database. Thereafter, the network of “active ingredient-targets” was constructed by cytoscape 3.7.2 software. The protein-protein interaction network was established by the BioGenet database to mine the potential protein function. Biological processes and pathways were analyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis. The binding ability of the core components of the Gleditsiae Spina and the core target of HGSC was verified by molecular docking and molecular dynamics simulation, and the therapeutic effect of Gleditsiae Spina was proved in vitro through cytotoxicity experiments. The effect of Gleditsiae Spina on the core pathway is obtained by western blotting. Results Gleditsiae Spina had cytotoxicity on HGSC based on network pharmacology and biological experiments. Luteolin, genistein, D-(+)-tryptophan, ursolic acid, and berberine are the identified core active ingredients of Gleditsiae Spina for regulating HGSC, with HPSE, PI3KCA, AKT1, and CTNNB1as the ideal targets. The prediction results were verified by molecular docking, molecular dynamic simulation, cell viability, and western blot analysis. Conclusion Gleditsiae Spina mainly downregulates the expression of heparanase and β-catenin to affect the composition of tumor cytoplasmic matrix and can regulate the PI3K-AKT pathway, integrating multiple targets and multiple pathways to play a therapeutic role. It also provides a theoretical basis for the prevention of ovarian cancer and its treatment using traditional Chinese medicine in the future.
Collapse
|
9
|
Cotoraci C, Ciceu A, Sasu A, Miutescu E, Hermenean A. Bioactive Compounds from Herbal Medicine Targeting Multiple Myeloma. APPLIED SCIENCES 2021; 11:4451. [DOI: 10.3390/app11104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple myeloma (MM) is one of the most widespread hematological cancers. It is characterized by a clonal proliferation of malignant plasma cells in the bone marrow and by the overproduction of monoclonal proteins. In recent years, the survival rate of patients with multiple myeloma has increased significantly due to the use of transplanted stem cells and of the new therapeutic agents that have significantly increased the survival rate, but it still cannot be completely cured and therefore the development of new therapeutic products is needed. Moreover, many patients have various side effects and face the development of drug resistance to current therapies. The purpose of this review is to highlight the bioactive active compounds (flavonoids) and herbal extracts which target dysregulated signaling pathway in MM, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their healing potential targeting multiple myeloma. Mechanistically, they demonstrated the ability to promote cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration/tumor progression, inhibition of angiogenesis in the tumor vascular network. Current research provides valuable new information about the ability of flavonoids to enhance the apoptotic effects of antineoplastic drugs, thus providing viable therapeutic options based on combining conventional and non-conventional therapies in MM therapeutic protocols.
Collapse
Affiliation(s)
- Coralia Cotoraci
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alina Ciceu
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alciona Sasu
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Eftimie Miutescu
- Department of Gastroenterology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
- Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| |
Collapse
|
10
|
Hussain Y, Luqman S, Meena A. Research Progress in Flavonoids as Potential Anticancer Drug Including Synergy with Other Approaches. Curr Top Med Chem 2021; 20:1791-1809. [PMID: 32357817 DOI: 10.2174/1568026620666200502005411] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND In chemotherapy for cancer, conventional drugs aim to target the rapidly growing and dividing cells at the early stages. However, at an advanced stage, cancer cells become less susceptible because of the multidrug resistance and the recruitment of alternative salvage pathways for their survival. Besides, owing to target non-selectivity, healthy proliferating cells also become vulnerable to the damage. The combination therapies offered using flavonoids to cure cancer not only exert an additive effect against cancer cells by targetting supplementary cell carnage pathways but also hampers the drug resistance mechanisms. Thus, the review aims to discuss the potential and pharmacokinetic limitations of flavonoids in cancer treatment. Further successful synergistic studies reported using flavonoids to treat cancer has been described along with potential drug delivery systems. METHODS A literature search was done by exploring various online databases like Pubmed, Scopus, and Google Scholar with the specific keywords like "Anticancer drugs", "flavonoids", "oncology research", and "pharmacokinetics". RESULTS Dietary phytochemicals, mainly flavonoids, hinder cell signalling responsible for multidrug resistance and cancer progression, primarily targeting cancer cells sparing normal cells. Such properties establish flavonoids as a potential candidate for synergistic therapy. However, due to low absorption and high metabolism rates, the bioavailability of flavonoids becomes a challenge. Such challenges may be overcome using novel approaches like derivatization, and single or co-delivery nano-complexes of flavonoids with conventional drugs. These new approaches may improve the pharmacokinetic and pharmacodynamic of flavonoids. CONCLUSION This review highlights the application of flavonoids as a potential anticancer phytochemical class in combination with known anti-cancer drugs/nanoparticles. It also discusses flavonoid's pharmacokinetics and pharmacodynamics issues and ways to overcome such issues. Moreover, it covers successful methodologies employed to establish flavonoids as a safe and effective phytochemical class for cancer treatment.
Collapse
Affiliation(s)
- Yusuf Hussain
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow-226015, Uttar Pradesh, India
| | - Suaib Luqman
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow-226015, Uttar Pradesh, India
| | - Abha Meena
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow-226015, Uttar Pradesh, India
| |
Collapse
|
11
|
Jiang Y, Fang Z, Leonard W, Zhang P. Phenolic compounds in Lycium berry: Composition, health benefits and industrial applications. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104340] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
12
|
Farooqi AA, Butt G, El-Zahaby SA, Attar R, Sabitaliyevich UY, Jovic JJ, Tang KF, Naureen H, Xu B. Luteolin mediated targeting of protein network and microRNAs in different cancers: Focus on JAK-STAT, NOTCH, mTOR and TRAIL-mediated signaling pathways. Pharmacol Res 2020. [DOI: https://doi.org/10.1016/j.phrs.2020.105188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Farooqi AA, Butt G, El-Zahaby SA, Attar R, Sabitaliyevich UY, Jovic JJ, Tang KF, Naureen H, Xu B. Luteolin mediated targeting of protein network and microRNAs in different cancers: Focus on JAK-STAT, NOTCH, mTOR and TRAIL-mediated signaling pathways. Pharmacol Res 2020; 160:105188. [PMID: 32919041 DOI: 10.1016/j.phrs.2020.105188] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
Abstract
There has always been a keen interest of basic and clinical researchers to search for cancer therapeutics having minimum off-target effects and maximum anticancer activities. In accordance with this approach, there has been an explosion in the field of natural products research in the past few decades because of extra-ordinary list of natural extracts and their biologically and pharmacologically active constituents having significant medicinal properties. Apparently, luteolin-mediated anticancer effects have been investigated in different cancers but there is superfluousness of superficial data. Generalized scientific evidence encompassing apoptosis, DNA damage and anti-inflammatory effects has been reported extensively. However, how luteolin modulates deregulated oncogenic pathways in different cancers has not been comprehensively uncovered. In this review we have attempted to focus on cutting-edge research which has unveiled remarkable abilities of luteolin to modulate deregulated oncogenic pathways in different cancers. We have partitioned the review into various sections to separately discuss advancements in therapeutic targeting of oncogenic protein networks. We have provided detailed mechanistic insights related to JAK-STAT signaling and summarized how luteolin inhibited STAT proteins to inhibit STAT-driven gene network. We have also individually analyzed Wnt/β-catenin and NOTCH pathway and how luteolin effectively targeted these pathways. Mapping of the signaling landscape has revealed that NOTCH pathway can be targeted therapeutically. NOTCH pathway was noted to be targeted by luteolin. We have also conceptually analyzed how luteolin restored TRAIL-induced apoptosis in resistant cancers. Luteolin induced an increase in pro-apoptotic proteins and efficiently inhibited anti-apoptotic proteins to induce apoptosis. Luteolin mediated regulation of non-coding RNAs is an exciting and emerging facet. Excitingly, there is sequential and systematic accumulation of clues which have started to shed light on intricate regulation of microRNAs by luteolin in different cancers. Collectively, sophisticated information will enable us to develop a refined understanding of the multi-layered regulation of signaling pathways and non-coding RNAs by luteolin in different cancers.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, 44000, Pakistan.
| | | | - Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University, Turkey
| | - Uteuliyev Yerzhan Sabitaliyevich
- Department of Health Policy and Health Care Development, Kazakh Medical University of Continuing Education, Almaty, 050004, Kazakhstan
| | - Jovana Joksimovic Jovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, SvetozaraMarkovića 69, 34000, Kragujevac, Serbia
| | - Kai-Fu Tang
- Digestive Cancer Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang, China
| | - Humaira Naureen
- Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, 519087, Guangdong, China.
| |
Collapse
|
14
|
Bi R, Deng Y, Tang C, Xuan L, Xu B, Du Y, Wang C, Wei W. Andrographolide sensitizes human renal carcinoma cells to TRAIL‑induced apoptosis through upregulation of death receptor 4. Oncol Rep 2020; 44:1939-1948. [PMID: 33000263 PMCID: PMC7551412 DOI: 10.3892/or.2020.7737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells, with minimal toxicity to normal tissues. However, accumulating evidence suggests that certain cancer types are insensitive to TRAIL signaling. The aim of this study was to identify an effective combination regimen, which can overcome TRAIL resistance in renal cancer cell. Herein, we found that human renal carcinoma cells (RCCs) are widely resistant to TRAIL-mediated growth inhibition and subsequently identified that andrographolide (Andro), a major constituent of Andrographis paniculate, an annual herbaceous plant in the family Acanthaceae, counteracts TRAIL resistance in RCCs. Combined treatment with TRAIL and Andro suppressed cell viability as determined by MTS and proliferation as determined by EdU in a dose-dependent manner and inactivated the clonogenic and migration ability of RCCs. Andro significantly enhances TRAIL-mediated cell cycle arrest at the G2/M phase as determined by flow cytometry and senescence. Moreover, Andro restored TRAIL signaling, which in turns activated pro-apoptosis caspases as determined by immunoblot assay. The TRAIL receptor, death receptor (DR)4, but not DR5, was found to be significantly upregulated in Andro-treated RCC cells, which contributed to the role of Andro as a TRAIL sensitizer. The present study demonstrated that the combined treatment of Andro and TRAIL has potential therapeutic value against renal cancer.
Collapse
Affiliation(s)
- Ran Bi
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuyou Deng
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chao Tang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Xuan
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bo Xu
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yujun Du
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Wei
- Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, Institute of Translational Medicine, Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| |
Collapse
|
15
|
Wu B, Xiong J, Zhou Y, Wu Y, Song Y, Wang N, Chen L, Zhang J. Luteolin enhances TRAIL sensitivity in non-small cell lung cancer cells through increasing DR5 expression and Drp1-mediated mitochondrial fission. Arch Biochem Biophys 2020; 692:108539. [PMID: 32777260 DOI: 10.1016/j.abb.2020.108539] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/21/2020] [Accepted: 08/02/2020] [Indexed: 12/28/2022]
Abstract
Cancer cells exhibit extreme sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) over normal cells, highlighting TRAIL's potential as a novel and effective cancer drug. However, the therapeutic effect of TRAIL is limited due to drug resistance. In the present study, we sought to investigate the potential effects of luteolin as a TRAIL sensitizer in non-small cell lung cancer (NSCLC) cells. A549 and H1975 cells had low sensitivity or were resistant to TRAIL. Luteolin alone or in combination with TRAIL decreased cell viability and increased apoptosis. Furthermore, luteolin alone or in combination with TRAIL enhanced death receptor 5 (DR5) expression and dynamin-related protein 1 (Drp1)-dependent mitochondrial fission. However, the synergistic effect of luteolin on cell viability and apoptosis was reversed by DR5 and Drp1 inhibition, suggesting that DR5 upregulation and mitochondrial dynamics may be essential for luteolin as a sensitizer of TRAIL-based therapy in NSCLC. Moreover, luteolin treatment alone or in combination with TRAIL increased the phosphorylation of c-Jun N-terminal kinase (JNK), while SP600125 (the JNK inhibitor) significantly abolished the synergistic effect on DR5 expression and Drp1 translocation, indicating that JNK signaling activation was greatly associated with the synergistic effect exerted by luteolin in NSCLC cells. Therefore, TRAIL combined with luteolin could be as an effective chemotherapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Bin Wu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jie Xiong
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yingtong Wu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yun Song
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ning Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lihua Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Jian Zhang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
16
|
Polonio-Alcalá E, Palomeras S, Torres-Oteros D, Relat J, Planas M, Feliu L, Ciurana J, Ruiz-Martínez S, Puig T. Fatty Acid Synthase Inhibitor G28 Shows Anticancer Activity in EGFR Tyrosine Kinase Inhibitor Resistant Lung Adenocarcinoma Models. Cancers (Basel) 2020; 12:cancers12051283. [PMID: 32438613 PMCID: PMC7281741 DOI: 10.3390/cancers12051283] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/29/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinases inhibitors (TKIs) are effective therapies for non-small cell lung cancer (NSCLC) patients whose tumors harbor an EGFR activating mutation. However, this treatment is not curative due to primary and secondary resistance such as T790M mutation in exon 20. Recently, activation of transducer and activator of transcription 3 (STAT3) in NSCLC appeared as an alternative resistance mechanism allowing cancer cells to elude the EGFR signaling. Overexpression of fatty acid synthase (FASN), a multifunctional enzyme essential for endogenous lipogenesis, has been related to resistance and the regulation of the EGFR/Jak2/STAT signaling pathways. Using EGFR mutated (EGFRm) NSCLC sensitive and EGFR TKIs’ resistant models (Gefitinib Resistant, GR) we studied the role of the natural polyphenolic anti-FASN compound (−)-epigallocatechin-3-gallate (EGCG), and its derivative G28 to overcome EGFR TKIs’ resistance. We show that G28’s cytotoxicity is independent of TKIs’ resistance mechanisms displaying synergistic effects in combination with gefitinib and osimertinib in the resistant T790M negative (T790M−) model and showing a reduction of activated EGFR and STAT3 in T790M positive (T790M+) models. Our results provide the bases for further investigation of G28 in combination with TKIs to overcome the EGFR TKI resistance in NSCLC.
Collapse
Affiliation(s)
- Emma Polonio-Alcalá
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, 17003 Girona, Spain;
| | - Sònia Palomeras
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
| | - Daniel Torres-Oteros
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain; (D.T.-O.); (J.R.)
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain; (D.T.-O.); (J.R.)
- Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Marta Planas
- LIPPSO, Department of Chemistry, University of Girona, 17003 Girona, Spain; (M.P.); (L.F.)
| | - Lidia Feliu
- LIPPSO, Department of Chemistry, University of Girona, 17003 Girona, Spain; (M.P.); (L.F.)
| | - Joaquim Ciurana
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, 17003 Girona, Spain;
| | - Santiago Ruiz-Martínez
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Correspondence: (S.R.-M.); (T.P.); Tel.: +34-972-419-548 (S.R.-M.); +34-972-419-628 (T.P.)
| | - Teresa Puig
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Correspondence: (S.R.-M.); (T.P.); Tel.: +34-972-419-548 (S.R.-M.); +34-972-419-628 (T.P.)
| |
Collapse
|
17
|
Chang CY, Li JR, Wu CC, Wang JD, Liao SL, Chen WY, Wang WY, Chen CJ. Endoplasmic Reticulum Stress Contributes to Indomethacin-Induced Glioma Apoptosis. Int J Mol Sci 2020; 21:ijms21020557. [PMID: 31952288 PMCID: PMC7013513 DOI: 10.3390/ijms21020557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
The dormancy of cellular apoptotic machinery has been highlighted as a crucial factor in therapeutic resistance, recurrence, and poor prognosis in patients with malignancy, such as malignant glioma. Increasing evidence indicates that nonsteroidal anti-inflammatory drugs (NSAIDs) confer chemopreventive effects, and indomethacin has been shown to have a novel chemotherapeutic application targeting glioma cells. To extend these findings, herein, we studied the underlying mechanisms of apoptosis activation caused by indomethacin in human H4 and U87 glioma cells. We found that the glioma cell-killing effects of indomethacin involved both death receptor- and mitochondria-mediated apoptotic cascades. Indomethacin-induced glioma cell apoptosis was accompanied by a series of biochemical changes, including reactive oxygen species generation, endoplasmic reticulum (ER) stress, apoptosis signal-regulating kinase-1 (Ask1) activation, p38 hyperphosphorylation, protein phosphatase 2A (PP2A) activation, Akt dephosphorylation, Mcl-1 and FLICE-inhibiting protein (FLIP) downregulation, Bax mitochondrial distribution, and caspases 3/caspase 8/caspase 9 activation. Data on pharmacological inhibition related to oxidative stress, ER stress, free Ca2+, and p38 revealed that the axis of oxidative stress/ER stress/Ask1/p38/PP2A/Akt comprised an apoptotic cascade leading to Mcl-1/FLIP downregulation and glioma apoptosis. Since indomethacin is an emerging choice in chemotherapy and its antineoplastic effects have been demonstrated in glioma tumor-bearing models, the findings further strengthen the argument for turning on the aforementioned axis in order to activate the apoptotic machinery of glioma cells.
Collapse
Affiliation(s)
- Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan;
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Financial Engineering, Providence University, Taichung City 433, Taiwan
- Department of Data Science and Big Data Analytics, Providence University, Taichung City 433, Taiwan
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City 407, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan;
| | - Wen-Yi Wang
- Department of Nursing, HungKuang University, Taichung City 433, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
- Correspondence: ; Tel.: +886-4-23592525 (ext. 4022)
| |
Collapse
|
18
|
Zeng Y, Li FD, Shi CW, Du JL, Xue YJ, Liu XY, Cao X, Wei N. Mechanism and therapeutic prospect of resveratrol combined with TRAIL in the treatment of renal cell carcinoma. Cancer Gene Ther 2019; 27:619-623. [PMID: 31664166 DOI: 10.1038/s41417-019-0150-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 11/09/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been reported to kill a wide variety of tumor cells with minimal effects on normal cell. However, renal cell carcinoma (RCC) cells 786-0 and OS-RC-2 were resistant to TRAIL. The present study examines the potential of combining polyphenolic compound resveratrol (RES) with TRAIL. We found that RES can sensitize RCC cells to TRAIL-induced death. Electron microscopy analyses showed that RES plus TRAIL can induce both autophagy and apoptosis in RCC cells. It was proved that the apoptosis is caspase-dependent and the activation of caspase-8, caspase-9, and caspase-3 was involved in this process. Besides, we also found that XIAP expression was significantly inhibited after RES plus TRAIL treatment in RCC cells. Furthermore, a fiber-modified replication-deficient adenovirus Ad5/35-TRAIL was generated to test the synergistic effect of RES and TRAIL in vivo. Our data demonstrated that RES plus Ad5/35-TRAIL significantly inhibited RCC xenograft growth in nude mice. These results suggest the possibility of a new combination therapeutic leading to the improvement of RCC treatment.
Collapse
Affiliation(s)
- Yan Zeng
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, 130118, Changchun, China
| | - Feng-di Li
- College of Life Science and Engineering, Center for Biomedical Research, Northwest Minzu University, 730030, Lanzhou, China
| | - Chun-Wei Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, 130118, Changchun, China
| | - Jiang-Long Du
- College of Life Science and Engineering, Center for Biomedical Research, Northwest Minzu University, 730030, Lanzhou, China
| | - Yu-Jia Xue
- College of Life Science and Engineering, Center for Biomedical Research, Northwest Minzu University, 730030, Lanzhou, China
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xin Cao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, 130118, Changchun, China. .,College of Life Science and Engineering, Center for Biomedical Research, Northwest Minzu University, 730030, Lanzhou, China.
| | - Na Wei
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China. .,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
19
|
Manzoor MF, Ahmad N, Ahmed Z, Siddique R, Zeng XA, Rahaman A, Muhammad Aadil R, Wahab A. Novel extraction techniques and pharmaceutical activities of luteolin and its derivatives. J Food Biochem 2019; 43:e12974. [PMID: 31489656 DOI: 10.1111/jfbc.12974] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/11/2019] [Accepted: 06/15/2019] [Indexed: 11/26/2022]
Abstract
Luteolin is a 3', 4', 5, 7 tetra hydroxyl flavonoid that exits in many plants, fruits, and vegetable. Many methods of extraction, isolation, and purification are being used, and therapeutic properties are being under discussion due to its valuable role in nutrition and human health. In this review, we have summarized conventional and novel extraction techniques from most recent research on luteolin, its derivatives, and its biological activities. Maceration, soxhlet, reflux, hydrodistillation, ultrasound-assisted extraction, microwave-assisted extraction, ultrasound microwave-assisted extraction, enzyme-assisted extraction, supercritical fluid extraction, and high-speed counter-current chromatography extraction techniques are being used for isolation and purification of these phytochemicals. The anti-inflammatory, anti-cancer, antioxidant, antiviral, heart protective, neurological impairments protection, anti-aging, and whiting properties have been discussed in this review. The literature suggests luteolin and its derivative has many promising health benefits and its therapeutic activity is strongly associated with isolating and purifying solvents and extraction techniques. PRACTICAL APPLICATIONS: This review aims to highlight the sources, novel extraction techniques, and pharmaceutical properties of luteolin. This review provides enough knowledge about how to get maximum extraction yield of luteolin using the novel extraction techniques. Because its therapeutic activity is strongly associated with isolating and purifying solvents and techniques.
Collapse
Affiliation(s)
- Muhammad Faisal Manzoor
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.,Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, China
| | - Nazir Ahmad
- Department of Food Science and Nutrition, Faculty of Life Science, Government College University, Faisalabad, Pakistan
| | - Zahoor Ahmed
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.,Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, China
| | - Rabia Siddique
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.,Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, China
| | - Abdul Rahaman
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.,Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, China
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Pakistan
| | - Abdul Wahab
- Department of Food Science and Nutrition, Faculty of Life Science, Government College University, Faisalabad, Pakistan
| |
Collapse
|
20
|
Fibronectin Promotes Cell Growth and Migration in Human Renal Cell Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20112792. [PMID: 31181623 PMCID: PMC6600362 DOI: 10.3390/ijms20112792] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Abstract
The prognostic and therapeutic values of fibronectin have been reported in patients with renal cell carcinoma (RCC). However, the underlying mechanisms of malignancy in RCC are not completely understood. We found that silencing of fibronectin expression attenuated human RCC 786-O and Caki-1 cell growth and migration. Silencing of potential fibronectin receptor integrin α5 and integrin β1 decreased 786-O cell ability in movement and chemotactic migration. Biochemical examination revealed a reduction of cyclin D1 and vimentin expression, transforming growth factor-β1 (TGF-β1) production, as well as Src and Smad phosphorylation in fibronectin-silenced 786-O and Caki-1 cells. Pharmacological inhibition of Src decreased 786-O cell growth and migration accompanied by a reduction of cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. In 786-O cells, higher activities in cell growth and migration than in Caki-1 cells were noted, along with elevated fibronectin and TGF-β1 expression. The additions of exogenous fibronectin and TGF-β1 promoted Caki-1 cell growth and migration, and increased cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. These findings highlight the role of fibronectin in RCC cell growth and migration involving Src and TGF-β1 signaling.
Collapse
|
21
|
Imran M, Rauf A, Abu-Izneid T, Nadeem M, Shariati MA, Khan IA, Imran A, Orhan IE, Rizwan M, Atif M, Gondal TA, Mubarak MS. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed Pharmacother 2019; 112:108612. [PMID: 30798142 DOI: 10.1016/j.biopha.2019.108612] [Citation(s) in RCA: 487] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022] Open
Abstract
Many food-derived phytochemicals and their derivatives represent a cornucopia of new anti-cancer compounds. Luteolin (3,4,5,7-tetrahydroxy flavone) is a flavonoid found in different plants such as vegetables, medicinal herbs, and fruits. It acts as an anticancer agent against various types of human malignancies such as lung, breast, glioblastoma, prostate, colon, and pancreatic cancers. It also blocks cancer development in vitro and in vivo by inhibition of proliferation of tumor cells, protection from carcinogenic stimuli, and activation of cell cycle arrest, and by inducing apoptosis through different signaling pathways. Luteolin can additionally reverse epithelial-mesenchymal transition (EMT) through a mechanism that involves cytoskeleton shrinkage, induction of the epithelial biomarker E-cadherin expression, and by down-regulation of the mesenchymal biomarkers N-cadherin, snail, and vimentin. Furthermore, luteolin increases levels of intracellular reactive oxygen species (ROS) by activation of lethal endoplasmic reticulum stress response and mitochondrial dysfunction in glioblastoma cells, and by activation of ER stress-associated proteins expressions, including phosphorylation of eIF2α, PERK, CHOP, ATF4, and cleaved-caspase 12. Accordingly, the present review article summarizes the progress of recent research on luteolin against several human cancers.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahor, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University of Science and Technology, Al Ain Campus, UAE
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari, Pakistan
| | - Mohammad Ali Shariati
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State, University Named After I.S. Turgenev, 302026, Orel, Russia
| | - Imtiaz Ali Khan
- Department of Agriculture, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan
| | - Ali Imran
- Department of Food Science, Nutrition & Home Economics, Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Muhammad Rizwan
- Department of Microbiology and Biotechnology, Abasyn University Peshawar, KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Australia
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
22
|
Ou YC, Li JR, Wang JD, Chen WY, Kuan YH, Yang CP, Liao SL, Lu HC, Chen CJ. Aspirin restores ABT-737-mediated apoptosis in human renal carcinoma cells. Biochem Biophys Res Commun 2018; 502:187-193. [PMID: 29792865 DOI: 10.1016/j.bbrc.2018.05.142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/19/2018] [Indexed: 02/01/2023]
Abstract
Aspirin is a novel chemopreventive agent against malignancy. However, outcomes of aspirin monotherapy of renal cell carcinoma (RCC) are inconsistent across studies. ABT-737, an BH3 mimetic inhibitor, is also a promising antitumor drug. Cancer cells including those from RCC, that have high levels of Mcl-1, are refractory to ABT-737-induced apoptosis. We here investigated how aspirin treatment modulates the ABT-737-induced apoptosis. Using the in vitro model of human 786-O cells, we showed that aspirin had sensitized cells to ABT-737 induced apoptosis. Such aspirin-induced changes of ABT-737 resistance was accompanied by a host of biochemical events like protein phosphatase 2A (PP2A) activation, AKT dephosphorylation, Mcl-1/FLICE inhibiting protein (FLIP)/XIAP downregulation, and Bax mitochondrial redistribution. The PP2A inhibitor, okadaic acid, was able to reverse the apirin-induced apoptotic changes. Apart from the aspirin treatment, Mcl-1 silencing also rendered cells vulnerable to ABT-737 induced apoptosis. Since PP2A, Akt, and Mcl-1 play critical roles in RCC malignancy and treatment resistance, our present study showed that aspirin, an alternative adjuvant agent, had recalled ABT-737 sensitivity in the RCC cells through processes involving the PP2A/Akt/Mcl-1 axis.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jiaan-Der Wang
- Department of Pediatrics & Child Health Care, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan
| | - Ching-Ping Yang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsi-Chi Lu
- Food Science Department and Graduate Institute, Tunghai University, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
23
|
Chang CY, Li JR, Wu CC, Wang JD, Yang CP, Chen WY, Wang WY, Chen CJ. Indomethacin induced glioma apoptosis involving ceramide signals. Exp Cell Res 2018; 365:66-77. [DOI: 10.1016/j.yexcr.2018.02.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 02/16/2018] [Accepted: 02/17/2018] [Indexed: 01/31/2023]
|
24
|
Liu T, Xu J, Yan HL, Cheng FC, Liu XJ. Luteolin Suppresses Teratoma Cell Growth and Induces Cell Apoptosis via Inhibiting Bcl-2. Oncol Res 2018. [PMID: 29540256 PMCID: PMC7848233 DOI: 10.3727/096504018x15208986577685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Luteolin, which is found in plant foods, has a range of therapeutic applications. In order to examine the potential roles of luteolin in ovarian teratocarcinoma, the human ovarian teratocarcinoma cell line PA-1 was selected for functional experiments in vitro and in vivo. We demonstrated that luteolin inhibited the proliferation and colony formation of PA-1 cells in vitro. The flow cytometry results suggested that luteolin induced apoptosis of PA-1 cells in a dose-dependent manner. Immunofluorescence and qRT-PCR results showed that the expression of B-cell lymphoma-2 (Bcl-2) was decreased in luteolin-treated cells, whereas the expression of Bcl-2-associated X (Bax) was increased compared with that in the control group. In addition, luteolin inhibited the tumor growth of ovarian teratocarcinoma cells in a xenograft model. All the results suggested that luteolin induced cell apoptosis and inhibited tumor growth of PA-1 cells.
Collapse
Affiliation(s)
- Teng Liu
- Department of Pediatric Surgery, Binzhou Medical University Hospital, Binzhou City, Shandong Province, P.R. China
| | - Juan Xu
- Pediatric Respiration and Intensive Care Unit, Binzhou Medical University Hospital, Shandong Province, P.R. China
| | - Hong Li Yan
- Electrocardiogram Room, Binzhou Medical University Hospital, Binzhou City, Shandong Province, P.R. China
| | - Feng Chun Cheng
- Department of Pediatric Surgery, Binzhou Medical University Hospital, Binzhou City, Shandong Province, P.R. China
| | - Xi Jie Liu
- Department of Pediatric Surgery, Binzhou Medical University Hospital, Binzhou City, Shandong Province, P.R. China
| |
Collapse
|
25
|
Lefranc F, Tabanca N, Kiss R. Assessing the anticancer effects associated with food products and/or nutraceuticals using in vitro and in vivo preclinical development-related pharmacological tests. Semin Cancer Biol 2017; 46:14-32. [PMID: 28602819 DOI: 10.1016/j.semcancer.2017.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
This review is part of a special issue entitled "Role of dietary pattern, foods, nutrients and nutraceuticals in supporting cancer prevention and treatment" and describes a pharmacological strategy to determine the potential contribution of food-related components as anticancer agents against established cancer. Therefore, this review does not relate to chemoprevention, which is analysed in several other reviews in the current special issue, but rather focuses on the following: i) the biological events that currently represent barriers against the treatment of certain types of cancers, primarily metastatic cancers; ii) the in vitro and in vivo pharmacological pre-clinical tests that can be used to analyse the potential anticancer effects of food-related components; and iii) several examples of food-related components with anticancer effects. This review does not represent a catalogue-based listing of food-related components with more or less anticancer activity. By contrast, this review proposes an original pharmacological strategy that researchers can use to analyse the potential anticancer activity of any food-related component-e.g., by considering the crucial characteristics of cancer biological aggressiveness. This review also highlights that cancer patients undergoing chemotherapy should restrict the use of "food complements" without supervision by a medical nutritionist. By contrast, an equilibrated diet that includes the food-related components listed herein would be beneficial for cancer patients who are not undergoing chemotherapy.
Collapse
Affiliation(s)
- Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles, 808 route de Lennik, 1070 Brussels, Belgium.
| | - Nurhayat Tabanca
- U.S Department of Agriculture-Agricultural Research Service, Subtropical Horticulture Research Station,13601 Old Cutler Rd., Miami, FL 33158, USA.
| | - Robert Kiss
- Retired-formerly at the Fonds National de la Recherche Scientifique (FRS-FNRS, Brussels, Belgium), 5 rue d'Egmont, 1000 Brussels, Belgium.
| |
Collapse
|
26
|
Novel Structurally Related Flavones Augment Cell Death Induced by rhsTRAIL. Int J Mol Sci 2017; 18:ijms18061211. [PMID: 28587286 PMCID: PMC5486034 DOI: 10.3390/ijms18061211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) was identified as a powerful activator of apoptosis in tumor cells and one of the most promising candidates for cancer therapy with no toxicity against normal tissues. However, many tumor cells are resistant to TRAIL-induced apoptosis. The aim of this work was to analyze the improvement of the anticancer effect of rhsTRAIL (recombinant human soluble TRAIL) by nine flavones: 5-Hydroxyflavone, 6-Hydroxyflavone, 7-Hydroxyflavone and their new synthetic derivatives 5-acetoxyflavone, 5-butyryloxyflavone, 6-acetoxyflavone, 6-butyryloxyflavone, 7-acetoxyflavone and 7-butyryloxyflavone. We examined the cytotoxic and apoptotic effects of rhsTRAIL enhanced by novel structurally-related flavones on SW480 and SW620 colon cancer cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test, the lactate dehydrogenase assay and annexin V-FITC fluorescence staining. We observed a slight difference in the activities of the flavones that was dependent on their chemical structure. Our study indicates that all nine flavones significantly augment cell death by rhsTRAIL (cytotoxicity range 36.8 ± 1.7%–91.4 ± 1.7%; apoptosis increase of 33.0 ± 0.7%–78.5 ± 0.9%). Our study demonstrates the potential use of tested flavones in TRAIL-based anticancer therapy and prevention.
Collapse
|
27
|
Luteolin selectively kills STAT3 highly activated gastric cancer cells through enhancing the binding of STAT3 to SHP-1. Cell Death Dis 2017; 8:e2612. [PMID: 28182003 PMCID: PMC5386483 DOI: 10.1038/cddis.2017.38] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 12/11/2022]
Abstract
The antitumor effect of luteolin, a plant flavonoid, in gastric cancer (GC) cells has not been fully understood. Here we show that luteolin selectively kills STAT3 overactivated GC cells that are often drug resistant. The treatment of luteolin in these GC cells significantly inhibited STAT3 phosphorylation and reduced the expression of STAT3 targeting gene Mcl-1, Survivin and Bcl-xl. Silencing of SHP-1, a protein tyrosine phosphatase, abolished the inhibitory effect of luteolin on STAT3 and cell apoptosis, suggesting that SHP-1 is crucial in luteolin-mediated cellular function. Moreover, this luteolin effect of STAT3 dephosphorylation by SHP-1 involved in HSP-90, which protected STAT3 phosphorylation by forming HSP-90/STAT3 complex. Thus, luteolin inhibited STAT3 activation through disrupting the binding of HSP-90 to STAT3, which promoted its interaction to SHP-1, resulted in the dephosphorylation of STAT3. The GC cell xenograft mouse model confirmed the effectiveness of luteolin induced inhibition of tumor growth in vivo.
Collapse
|
28
|
Epigallocatechin-3-gallate Sensitizes Human 786-O Renal Cell Carcinoma Cells to TRAIL-Induced Apoptosis. Cell Biochem Biophys 2016; 72:157-64. [PMID: 25539708 DOI: 10.1007/s12013-014-0428-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent. Epigallocatechin-3-gallate (EGCG) is a polyphenolic constituent of green tea. In this study, potentiating effect of EGCG on TRAIL-induced apoptosis human renal carcinoma cell line 786-O which is relatively resistant to TRAIL was examined, and the possible mechanism was investigated. Here, we show that co-treatment with EGCG and TRAIL induced significantly more profound apoptosis in 786-O cells. Treatment of 786-O cells with EGCG and TRAIL downregulated c-FLIP, Mcl-1, and Bcl-2 proteins in a caspase-dependent pathway. Moreover, we found that pretreatment with NAC markedly inhibited the expression levels of c-FLIP, Mcl-1, and Bcl-2 downregulated by the combinatory treatment, suggesting that the regulating effect of EGCG on these above apoptosis-relevant molecules was partially mediated by generation of ROS. Taken together, the present study demonstrates that EGCG sensitizes human 786-O renal cell carcinoma cells to TRAIL-induced apoptosis by downregulation of c-FLIP, Mcl-1, and Bcl-2.
Collapse
|
29
|
Abstract
Many food-derived phytochemical compounds and their derivatives represent a cornucopia of new anticancer compounds. Despite extensive study of luteolin, the literature has no information on the exact mechanisms or molecular targets through which it deters cancer progression. This review discusses existing data on luteolin's anticancer activities and then offers possible explanations for and molecular targets of its cancer-preventive action. Luteolin prevents tumor development largely by inactivating several signals and transcription pathways essential for cancer cells. This review also offers insights into the molecular mechanisms and targets through which luteolin either prevents cancer or mediates cancer cell death.
Collapse
|
30
|
Zhang JS, Lei JP, Wei GQ, Chen H, Ma CY, Jiang HZ. Natural fatty acid synthase inhibitors as potent therapeutic agents for cancers: A review. PHARMACEUTICAL BIOLOGY 2016; 54:1919-1925. [PMID: 26864638 DOI: 10.3109/13880209.2015.1113995] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/02/2015] [Accepted: 10/24/2015] [Indexed: 06/05/2023]
Abstract
Context Fatty acid synthase (FAS) is the only mammalian enzyme to catalyse the synthesis of fatty acid. The expression level of FAS is related to cancer progression, aggressiveness and metastasis. In recent years, research on natural FAS inhibitors with significant bioactivities and low side effects has increasingly become a new trend. Herein, we present recent research progress on natural fatty acid synthase inhibitors as potent therapeutic agents. Objective This paper is a mini overview of the typical natural FAS inhibitors and their possible mechanism of action in the past 10 years (2004-2014). Method The information was collected and compiled through major databases including Web of Science, PubMed, and CNKI. Results Many natural products induce cancer cells apoptosis by inhibiting FAS expression, with fewer side effects than synthetic inhibitors. Conclusion Natural FAS inhibitors are widely distributed in plants (especially in herbs and foods). Some natural products (mainly phenolics) possessing potent biological activities and stable structures are available as lead compounds to synthesise promising FAS inhibitors.
Collapse
Affiliation(s)
- Jia-Sui Zhang
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
| | - Jie-Ping Lei
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
| | - Guo-Qing Wei
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
| | - Hui Chen
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
| | - Chao-Ying Ma
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
- b School of Medicine, Southwest Jiaotong University , Chengdu , China
| | - He-Zhong Jiang
- a School of Life Science and Engineering, Southwest Jiaotong University , Chengdu , China
| |
Collapse
|
31
|
Kure A, Nakagawa K, Kondo M, Kato S, Kimura F, Watanabe A, Shoji N, Hatanaka S, Tsushida T, Miyazawa T. Metabolic Fate of Luteolin in Rats: Its Relationship to Anti-inflammatory Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:4246-4254. [PMID: 27170112 DOI: 10.1021/acs.jafc.6b00964] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Luteolin is a naturally occurring flavone that reportedly has anti-inflammatory effects. Because most luteolin is conjugated following intestinal absorption, free luteolin is likely present at low levels in the body. Therefore, luteolin metabolites are presumably responsible for luteolin bioactivity. Here we confirmed that luteolin glucuronides, especially luteolin-3'-O-glucuronide, are the major metabolites found in plasma after oral administration of luteolin (aglycone) or luteolin glucoside (luteolin-7-O-glucoside) to rats. Luteolin-4'-O-glucuronide and luteolin-7-O-glucuronide were also detectable together with luteolin-3'-O-glucuronide in the liver, kidney, and small intestine. Next, we prepared these luteolin glucuronides and compared the anti-inflammatory effects of luteolin and luteolin glucuronides on gene expression in lipopolysaccharide-treated RAW264.7 cells. Luteolin glucuronides, especially luteolin-7-O-glucuronide, reduced expression of inflammatory genes in the cells, although their effects were weaker than those of luteolin. These results indicate that the active compound responsible for the anti-inflammatory effect of luteolin in vivo would be luteolin glucuronide and/or residual luteolin.
Collapse
Affiliation(s)
- Ayako Kure
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| | - Momoko Kondo
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| | - Shunji Kato
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| | - Fumiko Kimura
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| | - Akio Watanabe
- Food Function Research Team, Saito Laboratory, Japan Food Research Laboratories , Ibaraki, Osaka 567-0085, Japan
| | - Naoki Shoji
- Miyagi Prefecture Watari Agricultural Promotion Center , Watari, Miyagi 989-2301, Japan
| | - Sakiko Hatanaka
- Industrial Technology Institute, Miyagi Prefectural Government , Sendai, Miyagi 981-3206, Japan
| | - Tojiro Tsushida
- Department of Food Management, School of Food, Agricultural and Environmental Sciences, Miyagi University , Sendai, Miyagi 982-0215, Japan
| | - Teruo Miyazawa
- Food and Biotechnology Innovation Project, New Industry Creation Hatchery Center (NICHe), Tohoku University , Sendai, Miyagi 980-8579, Japan
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University , Sendai, Miyagi 981-8555, Japan
| |
Collapse
|
32
|
Dia VP, Pangloli P. Epithelial-to-Mesenchymal Transition in Paclitaxel-Resistant Ovarian Cancer Cells Is Downregulated by Luteolin. J Cell Physiol 2016; 232:391-401. [PMID: 27198989 DOI: 10.1002/jcp.25436] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/18/2016] [Indexed: 12/21/2022]
Abstract
Ovarian cancer (OVCA) is the deadliest of all gynecological cancers which is attributed to late presentation, persistence, and development of chemoresistance. The objectives were to evaluate the association between OVCA paclitaxel-resistance and epithelial-to-mesenchymal transition (EMT) and to determine the capability of luteolin to chemosensitize OVCA cells. X10 and X22 cells were 11.8-25.3-fold and 7.8-8.6-fold resistant to paclitaxel than 1AP cells. X10 and X22 cells exhibited a mesenchymal phenotype, while 1AP has an epithelial characteristics. Furthermore, the expression of the epithelial marker E-cadherin was downregulated, while mesenchymal markers Vimentin and N-cadherin were upregulated in X10 and X22 cells when compared to 1AP cells. Transcription factors Snail, Slug, and Twist1 were upregulated in X10 cells, while Twist1 was highly expressed in X22 cells. Luteolin treatment caused cytotoxicity being most potent to X10 OVCA cells. Treatment of non-cytotoxic dose of luteolin at 15.625 μM chemosensitized X10 and X22 OVCA cells to paclitaxel as evidenced by reduced ED50 values from 11.8 to 0.2 μM and 8.6 to 3.6 μM for X10 and X22 cells, respectively. Moreover, luteolin treatment led to a more epithelial phenotype of X10 and X22 cells and modification of EMT markers indicating reversal of EMT. The mechanism involved is through reduction of phosphorylation of FAK and ERK leading to reduced nuclear translocation of p65. Our results highlight the significance of EMT in OVCA resistance to paclitaxel and warrant the investigation of luteolin as a potential therapeutic agent in chemoresistant OVCA. J. Cell. Physiol. 232: 391-401, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vermont P Dia
- Department of Food Science and Technology, University of Tennessee Institute of Agriculture, Knoxville, Tennessee.
| | - Philipus Pangloli
- Department of Food Science and Technology, University of Tennessee Institute of Agriculture, Knoxville, Tennessee
| |
Collapse
|
33
|
Dai X, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp Biol Med (Maywood) 2015; 240:760-73. [PMID: 25854879 DOI: 10.1177/1535370215579167] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to selectively induce apoptotic cell death in various tumor cells by engaging its death-inducing receptors (TRAIL-R1 and TRAIL-R2). This property has led to the development of a number of TRAIL-receptor agonists such as the soluble recombinant TRAIL and agonistic antibodies, which have shown promising anticancer activity in preclinical studies. However, besides activating caspase-dependent apoptosis in several cancer cells, TRAIL may also activate nonapoptotic signal transduction pathways such as nuclear factor-kappa B, mitogen-activated protein kinases, AKT, and signal transducers and activators of transcription 3, which may contribute to TRAIL resistance that is being now frequently encountered in various cancers. TRAIL resistance can be overcome by the application of efficient TRAIL-sensitizing pharmacological agents. Natural compounds have shown a great potential in sensitizing cells to TRAIL treatment through suppression of distinct survival pathways. In this review, we have summarized both apoptotic and nonapoptotic pathways activated by TRAIL, as well as recent advances in developing TRAIL-receptor agonists for cancer therapy. We also briefly discuss combination therapies that have shown great potential in overcoming TRAIL resistance in various tumors.
Collapse
Affiliation(s)
- Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingwen Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Frank Arfuso
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - M E Zayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
34
|
Yamamoto K, Shichiri H, Uda A, Yamashita K, Nishioka T, Kume M, Makimoto H, Nakagawa T, Hirano T, Hirai M. Apoptotic Effects of the Extracts of Cordyceps militaris
via Erk Phosphorylation in a Renal Cell Carcinoma Cell Line. Phytother Res 2015; 29:707-13. [DOI: 10.1002/ptr.5305] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/17/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Kazuhiro Yamamoto
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Hiroaki Shichiri
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Atsushi Uda
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Kazuhiko Yamashita
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Tatsuya Nishioka
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Manabu Kume
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Hiroo Makimoto
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Tsutomu Nakagawa
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Takeshi Hirano
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Midori Hirai
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| |
Collapse
|
35
|
Rana A, Attar R, Qureshi MZ, Gasparri ML, Donato VD, Ali GM, Farooqi AA. Dealing naturally with stumbling blocks on highways and byways of TRAIL induced signaling. Asian Pac J Cancer Prev 2014; 15:8041-6. [PMID: 25338981 DOI: 10.7314/apjcp.2014.15.19.8041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In-depth analysis of how TRAIL signals through death receptors to induce apoptosis in cancer cells using high throughput technologies has added new layers of knowledge. However, the wealth of information has also highlighted the fact that TRAIL induced apoptosis may be impaired as evidenced by experimental findings obtained from TRAIL resistant cancer cell lines. Overwhelmingly, increasing understanding of TRAIL mediated apoptosis has helped in identifying synthetic and natural compounds which can restore TRAIL induced apoptosis via functionalization of either extrinsic or intrinsic pathways. Increasingly it is being realized that biologically active phytochemicals modulate TRAIL induced apoptosis, as evidenced by cell-based studies. In this review we have attempted to provide an overview of how different phytonutrients have shown efficacy in restoring apoptosis in TRAIL resistant cancer cells. We partition this review into how the TRAIL mediated signaling landscape has broadened over the years and how TRAIL induced signaling machinery crosstalks with autophagic protein networks. Subsequently, we provide a generalized view of considerable biological activity of coumarins against a wide range of cancer cell lines and how coumarins (psoralidin and esculetin) isolated from natural sources have improved TRAIL induced apoptosis in resistant cancer cells. We summarize recent updates on piperlongumine, phenethyl isothiocyanate and luteolin induced activation of TRAIL mediated apoptosis. The data obtained from pre-clinical studies will be helpful in translation of information from benchtop to the bedside.
Collapse
Affiliation(s)
- Aamir Rana
- National Institute for Genomics and Advanced Biotechnology (NIGAB), NARC, Islamabad, Pakistan E-mail :
| | | | | | | | | | | | | |
Collapse
|
36
|
Juengel E, Nowaz S, Makarevi J, Natsheh I, Werner I, Nelson K, Reiter M, Tsaur I, Mani J, Harder S, Bartsch G, Haferkamp A, Blaheta RA. HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A. Mol Cancer 2014; 13:152. [PMID: 24935000 PMCID: PMC4073177 DOI: 10.1186/1476-4598-13-152] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Background Targeted therapies have improved therapeutic options of treating renal cell carcinoma (RCC). However, drug response is temporary due to resistance development. Methods Functional and molecular changes in RCC Caki-1 cells, after acquired resistance to the mammalian target of rapamycin (mTOR)-inhibitor everolimus (Cakires), were investigated with and without additional application of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA). Cell growth was evaluated by MTT assay, cell cycle progression and apoptosis by flow cytometry. Target molecules of everolimus and VPA, apoptotic and cell cycle regulating proteins were investigated by western blotting. siRNA blockade was performed to evaluate the functional relevance of the proteins. Results Everolimus resistance was accompanied by significant increases in the percentage of G2/M-phase cells and in the IC50. Akt and p70S6K, targets of everolimus, were activated in Cakires compared to drug sensitive cells. The most prominent change in Cakires cells was an increase in the cell cycle activating proteins cdk2 and cyclin A. Knock-down of cdk2 and cyclin A caused significant growth inhibition in the Cakires cells. The HDAC-inhibitor, VPA, counteracted everolimus resistance in Cakires, evidenced by a significant decrease in tumor growth and cdk2/cyclin A. Conclusion It is concluded that non-response to everolimus is characterized by increased cdk2/cyclin A, driving RCC cells into the G2/M-phase. VPA hinders everolimus non-response by diminishing cdk2/cyclin A. Therefore, treatment with HDAC-inhibitors might be an option for patients with advanced renal cell carcinoma and acquired everolimus resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Roman A Blaheta
- Department of Urology, Goethe-University, Interdisciplinary Science Building, Building 25A, Room 404, Theodor-Stern-Kai 7, Frankfurt / Main D-60590, Germany.
| |
Collapse
|