1
|
Norkaew C, Subkorn P, Chatupheeraphat C, Roytrakul S, Tanyong D. Pinostrobin, a fingerroot compound, regulates miR-181b-5p and induces acute leukemic cell apoptosis. Sci Rep 2023; 13:8084. [PMID: 37208425 DOI: 10.1038/s41598-023-35193-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/14/2023] [Indexed: 05/21/2023] Open
Abstract
Pinostrobin (PN) is the most abundant flavonoid found in fingerroot. Although the anti-leukemic properties of PN have been reported, its mechanisms are still unclear. MicroRNAs (miRNAs) are small RNA molecules that function in posttranscriptional silencing and are increasingly being used in cancer therapy. The aims of this study were to investigate the effects of PN on proliferation inhibition and induction of apoptosis, as well as the involvement of miRNAs in PN-mediated apoptosis in acute leukemia. The results showed that PN reduced cell viability and induced apoptosis in acute leukemia cells via both intrinsic and extrinsic pathways. A bioinformatics approach and Protein-Protein Interaction (PPI) network analysis revealed that ataxia-telangiectasia mutated kinase (ATM), one of the p53 activators that responds to DNA damage-induced apoptosis, is a crucial target of PN. Four prediction tools were used to predict ATM-regulated miRNAs; miR-181b-5p was the most likely candidate. The reduction in miR-181b-5 after PN treatment was found to trigger ATM, resulting in cellular apoptosis. Therefore, PN could be developed as a drug for acute leukemia; in addition, miR-181b-5p and ATM may be promising therapeutic targets.
Collapse
Affiliation(s)
- Chosita Norkaew
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Paweena Subkorn
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Chawalit Chatupheeraphat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology for Development Agency, Pathum Thani, 12120, Thailand
| | - Dalina Tanyong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
2
|
Naes SM, Ab-Rahim S, Mazlan M, Amir Hashim NA, Abdul Rahman A. Increased ENT2 expression and its association with altered purine metabolism in cell lines derived from different stages of colorectal cancer. Exp Ther Med 2023; 25:212. [PMID: 37123217 PMCID: PMC10133795 DOI: 10.3892/etm.2023.11911] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/21/2023] [Indexed: 05/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant cancer types worldwide. Although the purine metabolism pathway is vital for cancer cell survival, little is known about the role of equilibrative nucleoside transporter 2 (ENT2) in CRC development and its association with purine metabolites. The aim of the present study was to evaluate the levels of hypoxanthine phosphoribosyl transferase (HPRT), hypoxanthine and uric acid (UA), as well as xanthine oxidase (XO) activity, and investigate their association with ENT2 expression levels in a normal human colon cell line and CRC cell lines derived from different stages of CRC. These analyses were performed using the normal colon CCD-841CoN cell line and a panel of human CRC cell lines comprising SW480, HCT15 and HCT116, which represent Dukes' B, C and D stages, respectively. Reverse transcription-quantitative PCR was performed to determine the level of ENT2 mRNA expression. In cells of all CRC stages, the levels of HPRT and hypoxanthine were significantly higher (P<0.05), while XO activity and UA levels were significantly decreased (P<0.05), compared with those in the CCD-841CoN cell line. ENT2 expression was found to be elevated in cells derived from all stages of CRC. The Dukes' D stage cell line had higher levels of HPRT and hypoxanthine, although its ENT2 level was not significantly lower than that of the Dukes' B and C stage cell lines. Increased levels of HPRT and hypoxanthine in various stages of CRC may indicate an increase in the activity of the salvage pathway. The increased expression of ENT2 implies the importance of the ENT2 protein in facilitating hypoxanthine transport, which is required for enhanced DNA synthesis via hypoxanthine recycling. In conclusion, ENT2 may have potential as a target in the development of CRC therapeutics.
Collapse
Affiliation(s)
- Safaa M. Naes
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
- Institute of Medical and Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Sharaniza Ab-Rahim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Musalmah Mazlan
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Nurul Azmir Amir Hashim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Amirah Abdul Rahman
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
- Correspondence to: Dr Amirah Abdul Rahman, Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Selangor 47000, Malaysia
| |
Collapse
|
3
|
Dey S, Das A, Hossain MF. Galiellalactone: a review on synthetic strategies and tactics. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
4
|
Łuczkowska K, Kulig P, Baumert B, Machaliński B. The Evidence That 25(OH)D3 and VK2 MK-7 Vitamins Influence the Proliferative Potential and Gene Expression Profiles of Multiple Myeloma Cells and the Development of Resistance to Bortezomib. Nutrients 2022; 14:5190. [PMID: 36501221 PMCID: PMC9736786 DOI: 10.3390/nu14235190] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable hematological malignancy. Bortezomib (BTZ) is a proteasome inhibitor widely used in MM therapy whose potent activity is often hampered by the development of resistance. The immune system is vital in the pathophysiology of BTZ resistance. Vitamins D (VD) and K (VK) modulate the immune system; therefore, they are potentially beneficial in MM. The aim of the study was to evaluate the effect of BTZ therapy and VD and VK supplementation on the proliferation potential and gene expression profiles of MM cells in terms of the development of BTZ resistance. The U266 MM cell line was incubated three times with BTZ, VD and VK at different timepoints. Then, proliferation assays, RNA sequencing and bioinformatics analysis were performed. We showed BTZ resistance to be mediated by processes related to ATP metabolism and oxidative phosphorylation. The upregulation of genes from the SNORDs family suggests the involvement of epigenetic mechanisms. Supplementation with VD and VK reduced the proliferation of MM cells in both the non-BTZ-resistant and BTZ-resistant phenotypes. VD and VK, by restoring proper metabolism, may have overcome resistance to BTZ in vitro. This observation forms the basis for further clinical trials evaluating VD and VK as potential adjuvant therapies for MM patients.
Collapse
Affiliation(s)
- Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Piotr Kulig
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Bartłomiej Baumert
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252 Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252 Szczecin, Poland
| |
Collapse
|
5
|
Development and Validation of a Novel Circadian Rhythm-Related Signature to Predict the Prognosis of the Patients with Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4263261. [PMID: 35993051 PMCID: PMC9391189 DOI: 10.1155/2022/4263261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/06/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most important causes of cancer-related deaths and remains a major public health challenge worldwide. Considering the extensive heterogeneity of HCC, more accurate prognostic models are imperative. The circadian genes regulate the daily oscillations of key biological processes, such as nutrient metabolism in the liver. Circadian rhythm disruption has recently been recognized as an independent risk factor for cancer. In this study, The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) were compared and 248 differentially expressed genes (DEGs) of the circadian rhythm were identified. HCC was classified into two subtypes based on these DEGs. The prognostic value of each circadian rhythm-associated gene (CRG) for survival was assessed by constructing a multigene signature from TCGA cohort. A 6-gene signature was created by applying the least absolute shrinkage and selection operator (LASSO) Cox regression method, and all patients in TCGA cohort were divided into high- and low-risk groups according to their risk scores. The survival rate of patients with HCC in the low-risk group was significantly higher than that in the high-risk group (p < 0.001). The patients with HCC in the Gene Expression Omnibus (GEO) cohort were also divided into two risk subgroups using the risk score of TCGA cohort, and the overall survival time (OS) was prolonged in the low-risk group (p = 0.012). Based on the clinical characteristics, the risk score was an independent predictor of OS in the patients with HCC. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that multiple metabolic pathways, cell cycle, etc., were enhanced in the high-risk group. Using the metabolic pathway single-sample gene set enrichment analysis (ssGSEA), it was found that the metabolic pathways in the high- and low-risk groups between TCGA and GEO cohorts were altered essentially in the same way. In conclusion, the circadian genes play an important role in HCC metabolic rearrangements and can be further used to predict the prognosis the patients with HCC.
Collapse
|
6
|
Chimplee S, Smythe C, Tipmanee V, Sukrong S, Kanokwiroon K. Anticancer mechanism of 7-α-hydroxyfrullanolide on microtubules and computational prediction of its target binding in triple-negative breast cancer cells. PeerJ 2022; 10:e13508. [PMID: 35651747 PMCID: PMC9150694 DOI: 10.7717/peerj.13508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/06/2022] [Indexed: 01/17/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) responds poorly to the available drugs; thus, the mortality rate associated with TNBC remains high. 7-α-Hydroxyfrullanolide (7HF) possesses anticancer properties and arrests cells in the G2/M-phase via modulation of several proteins involved in the G2/M-phase transition, as well as the mitotic checkpoint in MDA-MB-468 (TNBC) cells. Microtubules (MTs) dynamically regulate cell division in the G2/M phase and are related to cancer cell stress response. However, antimitotic drug cytotoxicity to multiple cancer resistance developed in response to drugs are obstacles faced to date. Here, the activity and mechanism via which 7HF controls MTs dynamics was investigated in MDA-MB-468 cells. Methods 7HF uptake by MDA-MB-468 cells was assessed using spectrophotometry. The drug-like properties of 7HF were predicted using the Swiss-absorption, distribution, metabolism, and excretion (ADME) webtool. Then, the effect of 7HF treatment (6, 12, and 24 µM) on the dynamic arrangement of MTs was assessed for 1, 12, and 24 h using indirect immunofluorescence. Polymerization of α- and β-tubulin was assessed using different 7HF concentrations in a cell-free system for 1 h. Cell proliferation assay with bromodeoxyuridine plus propidium iodide staining and flow cytometry was performed at different 7HF concentrations and time points. The mechanism of action was assessed by detecting the expression of proteins, including Bub3, cyclin B1, p-Cdk1 (Tyr15), Rb, p-Rb (Ser780), Chk1, p-Chk1 (Ser345), Chk2, p-Chk2 (Ser516), and p-H2AX (Ser139), using western blotting. Molecular docking was used to predict the molecular interactions between 7HF and tubulins in MTs. Results We observed that 7HF was able to enter the MDA-MB-468 cells. The ADME webtool analysis predicted that it possesses the high passive permeation and gastrointestinal absorption properties of drugs. Various concentrations of 7HF disrupted the dynamic arrangement of spindle MTs by causing radial spindle array shrinkage and expansion of fibrous spindle density and radial array lengths in a time-dependent manner. 7HF reduced polymerization of α-, β-tubulin in dose-dependent manner. 7HF also triggered DNA damage response by inducing G2/M and G1 phase arrests in a concentration and time-dependent manner, which occurred due to the upregulation of Bub3, Chk1, p-Chk1 (Ser345), p-Cdk1 (Tyr15), and cyclin B1. According to molecular docking analysis, 7HF preferred to bind to β-tubulin over α-tubulin. The lactone, ketone, and hydroxyl groups of 7HF supported the 7HF-tubulin interactions. Hydrogen bonding with a hydrocarbon ring and salt bridge attractive forces were responsible for the binding versatility of 7HF. Conclusions This is the first study to investigate the molecular mechanism, MTs interacting sites, and the internalization and drug-like properties of 7HF in TNBC cells. The findings will be useful for developing 7HF-based treatment for patients with TNBC.
Collapse
Affiliation(s)
- Siriphorn Chimplee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Carl Smythe
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Suchada Sukrong
- Center of Excellence in DNA Barcoding of Thai Medicinal Plants, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kanyanatt Kanokwiroon
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
7
|
Yadav P, Mimansa, Kailasam K, Shanavas A. Nontoxic Metal-Free Visible Light-Responsive Carbon Nitride Quantum Dots Cause Oxidative Stress and Cancer-Specific Membrane Damage. ACS APPLIED BIO MATERIALS 2022; 5:1169-1178. [DOI: 10.1021/acsabm.1c01219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Pranjali Yadav
- Institute of Nano Science and Technology, Sector-81, Knowledge City, SAS Nagar, Punjab 140306, India
| | - Mimansa
- Institute of Nano Science and Technology, Sector-81, Knowledge City, SAS Nagar, Punjab 140306, India
| | - Kamalakannan Kailasam
- Institute of Nano Science and Technology, Sector-81, Knowledge City, SAS Nagar, Punjab 140306, India
| | - Asifkhan Shanavas
- Institute of Nano Science and Technology, Sector-81, Knowledge City, SAS Nagar, Punjab 140306, India
| |
Collapse
|
8
|
Zhang L, Dong L, Yang L, Luo Y, Chen F. MiR-27a-5p regulates acrylamide-induced mitochondrial dysfunction and intrinsic apoptosis via targeting Btf3 in rats. Food Chem 2022; 368:130816. [PMID: 34416489 DOI: 10.1016/j.foodchem.2021.130816] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
Acrylamide (AA), a potential carcinogen, is commonly formed in foods rich in carbohydrates at high heat. It is known that AA-induced mitochondrial dysfunction is responsible for its toxicity. Previously we found AA exposure increased miR-27a-5p expression in livers of SD rats. Here, the regulation mechanism of miR-27a-5p in mitochondrial dysfunction was investigated in rat liver cell lines (IAR20) and SD rats. The results showed that the overexpressed miR-27a-5p contributes to modulating mitochondrial dysfunction and Btf3 is identified as its target gene. The knockdown of Btf3 increases the cleaved PARP1 level and the phosphorylation of ATM and p53, which results in mitochondria-dependent apoptosis. Therefore, the miR-27a-5p-Btf3-ATM-p53 axis might play a vital role in the promotion of AA-induced cell apoptosis through disrupting mitochondrial structure and function. This would provide a potential target for the assessment and intervention of AA toxicity.
Collapse
Affiliation(s)
- Lujia Zhang
- College of Food Science & Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Li Dong
- College of Food Science & Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Liuqing Yang
- College of Food Science & Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Yinghua Luo
- College of Food Science & Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Fang Chen
- College of Food Science & Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
9
|
Canonico B, Cangiotti M, Montanari M, Papa S, Fusi V, Giorgi L, Ciacci C, Ottaviani MF, Staneva D, Grabchev I. Characterization of a fluorescent 1,8-naphthalimide-functionalized PAMAM dendrimer and its Cu(ii) complexes as cytotoxic drugs: EPR and biological studies in myeloid tumor cells. Biol Chem 2021; 403:345-360. [PMID: 34883001 DOI: 10.1515/hsz-2021-0388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/18/2021] [Indexed: 01/18/2023]
Abstract
The activity and interacting ability of a polyamidoamine (PAMAM) dendrimer modified with 4-N-methylpiperazine-1,8-naphthalimide units (termed D) and complexed by Cu(ii) ions, towards healthy and cancer cells were studied. Comparative electron paramagnetic resonance (EPR) studies of the Cu(ii)-D complex are presented: coordination mode, chemical structure, flexibility and stability of these complexes, in the absence and presence of myeloid cancer cells and peripheral blood mononuclear cells (PBMC). The interactions of Cu(ii) ions in the biological media at different equilibrium times were studied, highlighting different stability and interacting conditions with the cells. Furthermore, flow cytometry and confocal analysis, trace the peculiar properties of the dendrimers in PBMC and U937 cells. Indeed, a new probe (Fly) was used as a potential fluorescent tool for biological imaging of Cu(ii). The study highlights that dendrimer and, mainly, the Cu(ii) metallodendrimer are cytotoxic agents for the cells, specifically for U937 tumor cells, inducing mitochondrial dysfunction, ROS increase and lysosome involvement. The metallodendrimer shows antitumor selectivity, fewer affecting healthy PBMC, inducing a massive apoptotic cell death on U937 cells, in line with the high stability of this complex, as verified by EPR studies. The results underline the potentiality of this metallodendrimer to be used as anticancer drug.
Collapse
Affiliation(s)
- Barbara Canonico
- Department of Biomolecular Sciences (DISB), University of Urbino, I-61029 Urbino, Italy
| | - Michela Cangiotti
- Department of Pure and Applied Sciences (DiSPeA), University of Urbino, I-61029 Urbino, Italy
| | - Mariele Montanari
- Department of Biomolecular Sciences (DISB), University of Urbino, I-61029 Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences (DISB), University of Urbino, I-61029 Urbino, Italy
| | - Vieri Fusi
- Department of Pure and Applied Sciences (DiSPeA), University of Urbino, I-61029 Urbino, Italy
| | - Luca Giorgi
- Department of Pure and Applied Sciences (DiSPeA), University of Urbino, I-61029 Urbino, Italy
| | - Caterina Ciacci
- Department of Biomolecular Sciences (DISB), University of Urbino, I-61029 Urbino, Italy
| | | | - Desislava Staneva
- University of Chemical Technology and Metallurgy, BG-1756 Sofia, Bulgaria
| | - Ivo Grabchev
- Sofia University "St. Kliment Ohridski", Faculty of Medicine, BG-1407 Sofia, Bulgaria
| |
Collapse
|
10
|
Amewu RK, Sakyi PO, Osei-Safo D, Addae-Mensah I. Synthetic and Naturally Occurring Heterocyclic Anticancer Compounds with Multiple Biological Targets. Molecules 2021; 26:7134. [PMID: 34885716 PMCID: PMC8658833 DOI: 10.3390/molecules26237134] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023] Open
Abstract
Cancer is a complex group of diseases initiated by abnormal cell division with the potential of spreading to other parts of the body. The advancement in the discoveries of omics and bio- and cheminformatics has led to the identification of drugs inhibiting putative targets including vascular endothelial growth factor (VEGF) family receptors, fibroblast growth factors (FGF), platelet derived growth factors (PDGF), epidermal growth factor (EGF), thymidine phosphorylase (TP), and neuropeptide Y4 (NY4), amongst others. Drug resistance, systemic toxicity, and drug ineffectiveness for various cancer chemo-treatments are widespread. Due to this, efficient therapeutic agents targeting two or more of the putative targets in different cancer cells are proposed as cutting edge treatments. Heterocyclic compounds, both synthetic and natural products, have, however, contributed immensely to chemotherapeutics for treatments of various diseases, but little is known about such compounds and their multimodal anticancer properties. A compendium of heterocyclic synthetic and natural product multitarget anticancer compounds, their IC50, and biological targets of inhibition are therefore presented in this review.
Collapse
Affiliation(s)
- Richard Kwamla Amewu
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| | - Patrick Opare Sakyi
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
- Department of Chemical Sciences, School of Sciences, University of Energy and Natural Resources, Sunyani P.O. Box 214, Ghana
| | - Dorcas Osei-Safo
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| | - Ivan Addae-Mensah
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| |
Collapse
|
11
|
Kumar P, Chauhan A, Kumar M, Kuanr BK, Kundu A, Solanki R, Kapur MK. In vitro and in silico anticancer potential analysis of Streptomyces sp. extract against human lung cancer cell line, A549. 3 Biotech 2021; 11:254. [PMID: 33987071 PMCID: PMC8105467 DOI: 10.1007/s13205-021-02812-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/26/2021] [Indexed: 12/25/2022] Open
Abstract
During our previous investigation, bioactive compounds present in the extract of Streptomyces sp. strain 196 were characterized using LC-MS/MS and 1H NMR studies. These compounds were K-252-C aglycone indolocarbazole alkaloid, decoyinine, and cycloheximide; the study of these natural drugs against lung carcinoma is still limited. Focus of the current investigation was to study the anticancer effect of strain 196 extract on lung cancer cells (A549). During in vitro studies, anti-proliferative effect of extract was studied using MTT assay in A549 cells. Effect of extract on cell survival was further evaluated using colony assay. Cell death was qualitatively assessed using apoptosis assay. The aftereffect of extract treatment on metastatic potential of cancerous cells was studied using wound closure assay. Effect of extract on the morphology and cytoskeletal arrangement of A549 cells was studied using phalloidin staining. The extract demonstrated concentration and time-dependent cytotoxicity with IC50 value at 0.5 mg/ml (6 h) and 0.15 mg/ml (24 h). The proliferation and metastatic potential of cells, as characterized by MTT and migration assay, decreased over time in a concentration-dependent manner. Discrete changes in cellular morphology were noted as a result of the induced cytotoxicity. Apoptosis assay demonstrated 98.7% cell death at highest concentration of extract (1 mg/ml). During in silico studies, molecular docking revealed that strain 196 compounds are efficiently binding to mutant EGFR form (T790M/L858R) with release of binding energy (∆G) between - 5 and - 6.9 kcal/Mol. In conclusion, strain 196 extract could be a source of therapeutic drugs to treat lung carcinoma.
Collapse
Affiliation(s)
- Prateek Kumar
- Microbial Technology Lab, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110 019 India
| | - Anjali Chauhan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067 India
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Munendra Kumar
- Microbial Technology Lab, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110 019 India
| | - Bijoy K. Kuanr
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Aditi Kundu
- Division of Agricultural Chemicals, ICAR- Indian Agricultural Research Institute, New Delhi, 110012 India
| | - Renu Solanki
- Deen Dayal Upadhyaya College, University of Delhi, New Delhi, 110078 India
| | - Monisha Khanna Kapur
- Microbial Technology Lab, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110 019 India
| |
Collapse
|
12
|
Fadaka AO, Bakare OO, Sibuyi NRS, Klein A. Gene Expression Alterations and Molecular Analysis of CHEK1 in Solid Tumors. Cancers (Basel) 2020; 12:cancers12030662. [PMID: 32178478 PMCID: PMC7139733 DOI: 10.3390/cancers12030662] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/16/2022] Open
Abstract
Alterations in the Checkpoint kinase (CHEK1) gene, its regulation, and the possible clinical outcomes in human solid tumors have not been previously examined. Therefore, the present study was carried out to evaluate the expression of CHEK1 in solid tumors as well as the mechanism by which it can be regulated through non-coding RNAs. The expression of CHEK1 was investigated using Oncomine analysis. cBioPortal, Kaplan-Meier Plotter, and PrognoScan were performed to identify the prognostic roles of this gene in solid tumors. The copy number alteration, mutation, interactive analysis, and visualization of the altered networks were performed by cBioPortal. The molecular binding analysis was carried out by Schrodinger suite, PATCHDOCK, and discovery studio visualizer. The study demonstrated that the CHEK1 gene was differentially expressed in four different cancers, and that reduced CHEK1 mRNA expression is an unfavorable prognostic factor for patients with gastric and colorectal cancer. The molecular docking results showed that the CHEK1 gene can be regulated by microRNAs (miR-195-5p) due to the number of stable hydrogen atoms observed within the distance of 2.0 Å and the favorable amino acids (Ala221, Ile353, Ile365, Ile756, Val797, Val70, Val154, Ile159, Val347, Tyr804, Phe811, Tyr815, and Phe156) identified in the binding pocket of the argonaute protein. Due to the possibility of CHEK1's involvement in solid tumors, it may potentially be a target for therapeutic intervention in cancer. Further studies into the interaction between CHEK1 and other co-expressed genes may give further insight into other modes of regulation of this gene in cancer patients.
Collapse
Affiliation(s)
- Adewale Oluwaseun Fadaka
- Bioinformatics research group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
- Correspondence: ; Tel.: +27-630511928 or +234-8039242052
| | - Olalekan Olanrewaju Bakare
- Bioinformatics research group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa
| | - Ashwil Klein
- Plant Omics group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| |
Collapse
|
13
|
Ko H, Lee JH, Kim HS, Kim T, Han YT, Suh YG, Chun J, Kim YS, Ahn KS. Novel Galiellalactone Analogues Can Target STAT3 Phosphorylation and Cause Apoptosis in Triple-Negative Breast Cancer. Biomolecules 2019; 9:biom9050170. [PMID: 31058868 PMCID: PMC6571922 DOI: 10.3390/biom9050170] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/24/2022] Open
Abstract
Aberrant activation of signal transducer and activator of transcription 3 (STAT3) has been documented in various malignancies including triple-negative breast cancers (TNBCs). The STAT3 transcription factor can regulate the different important hallmarks of tumor cells, and thus, targeting it can be a potential strategy for treating TNBC, for which only limited therapeutic options are available. In this study, we analyzed the possible effect of (-)-galiellalactone and its novel analogues, SG-1709 and SG-1721, and determined whether these agents exerted their antineoplastic effects by suppressing the STAT3 signaling pathway in TNBC cells. The two analogues, SG-1709 and SG-1721, inhibited both constitutive as well as inducible STAT3 phosphorylation at tyrosine 705 more effectively than (-)-galiellalactone, which indicates that the analogues are more potent STAT3 blockers. Moreover, SG-1721 not only inhibited nuclear translocation and DNA binding of STAT3 but also induced apoptosis, and decreased expression of diverse oncogenic proteins. Interestingly, SG-1721 also exhibited an enhanced apoptotic effect when combined with radiotherapy. Furthermore, in vivo administration of SG-1721 significantly attenuated breast xenograft tumor growth via decreasing levels of p-STAT3. Therefore, SG-1721 may be a promising candidate for further application as a pharmacological agent that can target STAT3 protein in treating TNBC.
Collapse
Affiliation(s)
- Hyejin Ko
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Jong Hyun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Hyun Su Kim
- College of Pharmacy, CHA University, 120 Haeryong-ro, Pochen-si, Gyenggi-do 11160, Korea.
| | - Taewoo Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Young Taek Han
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 330-714, Korea.
| | - Young-Ger Suh
- College of Pharmacy, CHA University, 120 Haeryong-ro, Pochen-si, Gyenggi-do 11160, Korea.
| | - Jaemoo Chun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
14
|
Soubéran A, Cappaï J, Chocry M, Nuccio C, Raujol J, Colin C, Lafitte D, Kovacic H, Quillien V, Baeza-Kallee N, Rougon G, Figarella-Branger D, Tchoghandjian A. Inhibitor of Apoptosis Proteins Determines Glioblastoma Stem-Like Cell Fate in an Oxygen-Dependent Manner. Stem Cells 2019; 37:731-742. [PMID: 30920104 DOI: 10.1002/stem.2997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
In glioblastomas, apoptosis inhibitor proteins (IAPs) are involved in apoptotic and nonapoptotic processes. We previously showed that IAP inhibition induced a loss of stemness and glioblastoma stem cells differentiation by activating nuclear factor-κB under normoxic conditions. Hypoxia has been shown to modulate drug efficacy. Here, we investigated how IAPs participate in glioblastoma stem-like cell maintenance and fate under hypoxia. We showed that in a hypoxic environment, IAPs inhibition by GDC-0152, a small-molecule IAPs inhibitor, triggered stem-like cell apoptosis and decreased proliferation in four human glioblastoma cell lines. We set up a three-dimensional glioblastoma spheroid model in which time-of-flight secondary ion mass spectrometry analyses revealed a decrease in oxygen levels between the periphery and core. We observed low proliferative and apoptotic cells located close to the hypoxic core of the spheres and glial fibrillary acidic protein+ cells at their periphery. These oxygen-dependent GDC-0152 antitumoral effects have been confirmed on human glioblastoma explants. Notably, serine-threonine kinase activation analysis revealed that under hypoxic conditions, IAP inhibition activated ataxia telangiectasia and Rad3-related protein signaling. Our findings provide new insights into the dual mechanism of action of IAP inhibitors that depends on oxygen level and are relevant to their therapeutic application in tumors. Stem Cells 2019;37:731-742.
Collapse
Affiliation(s)
- Aurélie Soubéran
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Jessica Cappaï
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Mathieu Chocry
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | | | | | - Carole Colin
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Daniel Lafitte
- Aix-Marseille University, INSERM UMR MD1, Marseille, France
| | - Hervé Kovacic
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Véronique Quillien
- Rennes 1 University, "Chemistry, Oncogenesis, Stress, Signaling", INSERM U1242, Rennes, France.,Centre de lutte contre le cancer Eugène Marquis, Rennes F-35042, France
| | - Nathalie Baeza-Kallee
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Geneviève Rougon
- Aix-Marseille University, Institut de Neurosciences Timone, CNRS 7289, Marseille, France
| | - Dominique Figarella-Branger
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,AP-HM, Timone Hospital, Department of Anatomopathology, Marseille, France
| | - Aurélie Tchoghandjian
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| |
Collapse
|
15
|
Lu Y, Zhao S, Zhou S, Chen SC, Luo T. Enantioselective syntheses and application of 4-epi-galiellalactone and the corresponding activity-based probe: from strained bicycles to strained tricycles. Org Biomol Chem 2019; 17:1886-1892. [PMID: 30183048 DOI: 10.1039/c8ob01915k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The [6,5,5] tricyclic fungal metabolite galiellalactone is a Michael acceptor that has been demonstrated to be a covalent inhibitor for Signal Transducer and Activator of Transcription 3 (STAT3). Recognizing the ring strain associated with the skeleton of this natural product, we utilized 1R-5S-bicyclo[3.1.0]hexan-2-one as the starting material and developed two novel approaches to accomplish the enantioselective total synthesis of the C4 epimer of galiellalactone in 5 and 7 steps, respectively, which capitalized on an efficient radical cyclization/fragmentation cascade reaction. Furthermore, an activity-based probe of 4-epi-galiellalactone with a terminal alkyne tag was successfully prepared to enable the experiments of activity-based protein profiling (ABPP). Through western blot and proteomic analysis, we not only confirmed the known target STAT3, but also identified a new target protein ataxin-7, which formed a covalent bond with the probe in intact cells via the Cys-129 residue.
Collapse
Affiliation(s)
- Yandong Lu
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Ministry of Education, Beijing National Laboratory for Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | | | | | | | | |
Collapse
|
16
|
SWATH proteomic profiling of prostate cancer cells identifies NUSAP1 as a potential molecular target for Galiellalactone. J Proteomics 2019; 193:217-229. [DOI: 10.1016/j.jprot.2018.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022]
|
17
|
Next-Generation Sequencing Reveals the Role of Epigallocatechin-3-Gallate in Regulating Putative Novel and Known microRNAs Which Target the MAPK Pathway in Non-Small-Cell Lung Cancer A549 Cells. Molecules 2019; 24:molecules24020368. [PMID: 30669618 PMCID: PMC6359307 DOI: 10.3390/molecules24020368] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/31/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer constitutes 85% of non-small cell lung cancer diagnosed cases. MicroRNAs are novel biomarkers that are capable of modulating multiple oncogenic pathways. Epigallocatechin-3-gallate (EGCG) is a potent chemopreventive and chemotherapeutic agent for cancer. We aimed to identify important known and putative novel microRNAs modulated by EGCG in A549 cells using next-generation sequencing and identify their gene targets. Preliminary analysis revealed an IC50 value of 309 μM with G0/G1 phase arrest at 40 μM EGCG treatment. MicroRNA profiling identified 115 known and 4 putative novel microRNAs in 40 μM and 134 known and 3 putative novel microRNAs in 100 μM EGCG-treated A549 cells. The top 10 up-expressed microRNAs were similar between the untreated control and EGCG-treated A549 cells. An up-expression in oncogenic microRNAs, which belong to broadly conserved seed families, were observed in untreated control and EGCG-treated A549 cells. Kyoto Encyclopedia of Genes and Genomes and Protein Analysis Through Evolutionary Relationships pathway analyses of the validated microRNA targeting genes strengthened the hypothesis that EGCG treatment can modulate microRNAs that play a significant role in the MAPK signaling pathway. Expression profile of microRNAs was validation by quantitative real time PCR of randomly selected microRNAs. This study identified signature microRNAs that can be used as novel biomarkers for lung cancer diagnosis.
Collapse
|
18
|
Two decades of research in discovery of anticancer drugs targeting STAT3, how close are we? Pharmacol Ther 2018; 191:74-91. [DOI: 10.1016/j.pharmthera.2018.06.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
19
|
Wang J, Zhang YS, Thakur K, Hussain SS, Zhang JG, Xiao GR, Wei ZJ. Licochalcone A from licorice root, an inhibitor of human hepatoma cell growth via induction of cell apoptosis and cell cycle arrest. Food Chem Toxicol 2018; 120:407-417. [DOI: 10.1016/j.fct.2018.07.044] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 01/20/2023]
|
20
|
Pei C, He Q, Liang S, Gong X. Mahanimbine Exerts Anticancer Effects on Human Pancreatic Cancer Cells by Triggering Cell Cycle Arrest, Apoptosis, and Modulation of AKT/Mammalian Target of Rapamycin (mTOR) and Signal Transducer and Activator of Transcription 3 (STAT3) Signalling Pathways. Med Sci Monit 2018; 24:6975-6983. [PMID: 30273298 PMCID: PMC6178883 DOI: 10.12659/msm.911013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Pancreatic cancer causes tremendous mortality across the globe mainly due to late diagnosis and unavailability of efficient chemotheruptic agents. In the current study the anticancer potential of a plant derived alkaloid, Mahanimbine, was examined against a panel of pancreatic cancer cells. Material/Methods The cell proliferation was determined by MTT assay. Annexin V/PI and DAPI staining were performed to detect apoptosis. Cell cycle distribution was investigated by flow cytometery. Cell migration was detected by wound healing assay and protein expression was checked by western blotting. Results The results revealed that Mahanimbine could inhibit the proliferation of the all the pancreatic cancer cells with lower cytoxicity against the normal cells. The IC50 ranged from 3.5 to 64 μM against the pancreatic cancer cell lines. The lowest IC50 of 3.5 μM was observed tor the Capan-2 and SW119 pancreatic cancer cell lines. The anticancer activity of Mahanimbine against the Capan-2 and SW119 cells was found to be due to G0/G1 cell cycle arrest and induction of apoptosis. Mahanimbine prompted apoptosis was also associated with decline in Bcl-2 and enhancement of the Bax expression. Further, it was observed that Mahanimbine could inhibit the AKT/mTOR and STAT3 signalling pathways in the Capan-2 and SW119 pancreatic cancer cells. The effects of the Mahanimbine were also examined on the migration of the Capan-2 and SW119 pancreatic cancer cells. It was found that Mahanimbine could inhibit the motility and migration of both the pancreatic cancer cell lines. Conclusions We found that Mahanimbine inhibits the proliferation of pancreatic cancer cells and as such Mahanimbine may prove beneficial in the management of pancreatic cancer.
Collapse
Affiliation(s)
- Chenlin Pei
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Qun He
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Shuai Liang
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Xuejun Gong
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
21
|
Cui S, Wang Y, Gong Y, Lin X, Zhao Y, Zhi D, Zhou Q, Zhang S. Correlation of the cytotoxic effects of cationic lipids with their headgroups. Toxicol Res (Camb) 2018; 7:473-479. [PMID: 30090597 PMCID: PMC6062336 DOI: 10.1039/c8tx00005k] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
As effective non-viral vectors of gene therapy, cationic lipids still have the problem of toxicity, which has become one of the main bottlenecks for their applications. The toxicity of cationic lipids is strongly connected to the headgroup structures. In this article, we studied the cytotoxicity of two cationic lipids with a quaternary ammonium headgroup (CDA14) and a tri-peptide headgroup (CDO14), respectively, and with the same linker bond and hydrophobic domain. The IC50 values of CDA14 and CDO14 against NCI-H460 cells were 109.4 μg mL-1 and 340.5 μg mL-1, respectively. To determine the effects of headgroup structures of cationic lipids on cytotoxicity, apoptosis related pathways were investigated. As the lipids with a quaternary ammonium headgroup could induce more apoptotic cells than the ones with a peptide headgroup, the enzymatic activity of caspase-9 and caspase-3 increased obviously, whereas the mitochondrial membrane potential (MMP) decreased. At the same time, the reactive oxygen species (ROS) levels also increased and the cell cycle was arrested at the S phase. The results showed that the toxicity of the cationic lipid had a close relationship with its headgroup structures, and the cytotoxic mechanism was mainly via the caspase activation dependent signaling pathway and mitochondrial dysfunction. Through this study, we hope to provide the scientific basis for exploiting safer and more efficient cationic lipids for gene delivery.
Collapse
Affiliation(s)
- Shaohui Cui
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Yueying Wang
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Yan Gong
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Xiao Lin
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Defu Zhi
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Quan Zhou
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization , Ministry of Education , College of Life Science , Dalian Minzu University , Dalian 116600 , China .
| |
Collapse
|
22
|
Lee CC, Hsieh TS. Wuho/WDR4 deficiency inhibits cell proliferation and induces apoptosis via DNA damage in mouse embryonic fibroblasts. Cell Signal 2018; 47:16-26. [PMID: 29574139 DOI: 10.1016/j.cellsig.2018.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
Abstract
Wuho known as WDR4 encodes a highly conserved WD40-repeat protein, which has known homologues of WDR4 in human and mouse. Wuho-FEN1 interaction may have a critical role in the growth and development, and in the maintenance of genome stability. However, how Wuho gene deletion contributes to cell growth inhibition and apoptosis is still unknown. We utilized CAGGCre-ER transgenic mice have a tamoxifen-inducible cre-mediated recombination cassette to prepare primary mouse embryonic fibroblasts (MEFs) with Wuho deficiency. We have demonstrated that Wuho deficiency would induces γH2AX protein level elevation, heterochromatin relaxation and DNA damage down-stream sequences, including p53 activation, caspase-mediated apoptotic pathway, and p21-mediated G2/M cell cycle arrest.
Collapse
Affiliation(s)
- Chi-Chiu Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Sec. 2, Nangang, Taipei 11529, Taiwan.
| | - Tao-Shih Hsieh
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Sec. 2, Nangang, Taipei 11529, Taiwan; Department of Biochemistry, Duke University, Durham, NC, United States
| |
Collapse
|
23
|
Ramalingam V, Varunkumar K, Ravikumar V, Rajaram R. Target delivery of doxorubicin tethered with PVP stabilized gold nanoparticles for effective treatment of lung cancer. Sci Rep 2018; 8:3815. [PMID: 29491463 PMCID: PMC5830607 DOI: 10.1038/s41598-018-22172-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/15/2018] [Indexed: 02/06/2023] Open
Abstract
Development of drug delivery system conjugated with doxorubicin (dox) on the surface of AuNPs with polyvinylpyrrolidone (Dox@PVP-AuNPs), we have demonstrated that human lung cancer cells can significantly overcome by the combination of highly effective cellular entry and responsive intracellular release of doxorubicin from Dox@PVP-AuNPs complex. Previously drug release from doxorubicin-conjugated AuNPs was confirmed by the recovered fluorescence of doxorubicin from quenching due to the nanosurface energy transfer between doxorubicinyl groups and AuNPs. Dox@PVP-AuNPs achieved enhanced inhibition of lung cancer cells growth than free Doxorubicin and PVP-AuNPs. The in vitro cytotoxic effect of PVP-AuNPs, free Dox and Dox@PVP-AuNPs inhibited the proliferation of human lung cancer cells with IC50 concentration. Compared with control cells, PVP-AuNPs and free Dox, Dox@PVP-AuNPs can increases ROS generation, sensitize mitochondrial membrane potential and induces both early and late apoptosis in lung cancer cells. Moreover, Dox@PVP-AuNPs highly upregulates the expression of tumor suppressor genes than free Dox and PVP-AuNPs and induces intrinsic apoptosis in lung cancer cells. From the results, Dox@PVP-AuNPs can be considered as an potential drug delivery system for effective treatment of human lung cancer.
Collapse
Affiliation(s)
- Vaikundamoorthy Ramalingam
- DNA Barcoding and Marine Genomics lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620 024, Tamil Nadu, India
| | - Krishnamoorthy Varunkumar
- Cancer Biology Lab, Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620 024, Tamil Nadu, India
| | - Vilwanathan Ravikumar
- Cancer Biology Lab, Department of Biochemistry, Bharathidasan University, Tiruchirappalli, 620 024, Tamil Nadu, India
| | - Rajendran Rajaram
- DNA Barcoding and Marine Genomics lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620 024, Tamil Nadu, India.
| |
Collapse
|
24
|
Shu G, Zhang L, Jiang S, Cheng Z, Wang G, Huang X, Yang X. Isoliensinine induces dephosphorylation of NF-kB p65 subunit at Ser536 via a PP2A-dependent mechanism in hepatocellular carcinoma cells: roles of impairing PP2A/I2PP2A interaction. Oncotarget 2018; 7:40285-40296. [PMID: 27244888 PMCID: PMC5130008 DOI: 10.18632/oncotarget.9603] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/26/2016] [Indexed: 01/04/2023] Open
Abstract
Our previous study discovered that isoliensinine (isolie) triggers hepatocellular carcinoma (HCC) cell apoptosis via inducing p65 dephosphorylation at Ser536 and inhibition of NF-κB. Here, we showed that isolie promoted p65/PP2A interaction in vitro and in vivo. Repression of PP2A activity or knockdown of the expression of PP2A-C (the catalytic subunit of PP2A) abrogated isolie-provoked p65 dephosphorylation. I2PP2A is an endogenous PP2A inhibitor. Isolie directly impaired PP2A/I2PP2A interaction. Knockdown of I2PP2A boosted p65/PP2A association and p65 dephosphorylation. Overexpression of I2PP2A restrained isolie-induced p65 dephosphorylation. Untransformed hepatocytes were insensitive to isolie-induced NF-κB inhibition and cell apoptosis. In these cells, basal levels of I2PP2A and p65 phosphorylation at Ser536 were lower than in HCC cells. These findings collectively indicated that isolie suppresses NF-κB in HCC cells through impairing PP2A/I2PP2A interaction and stimulating PP2A-dependent p65 dephosphorylation at Ser536.
Collapse
Affiliation(s)
- Guangwen Shu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Lang Zhang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Shanqing Jiang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Zhuo Cheng
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Guan Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Xu Huang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, PR China
| |
Collapse
|
25
|
Cho HD, Lee JH, Moon KD, Park KH, Lee MK, Seo KI. Auriculasin-induced ROS causes prostate cancer cell death via induction of apoptosis. Food Chem Toxicol 2018; 111:660-669. [PMID: 29217266 DOI: 10.1016/j.fct.2017.12.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 01/11/2023]
Abstract
Recent studies have demonstrated that natural agents targeting the accumulation of reactive oxygen species (ROS) that selectively kill, leaving normal cells undamaged, can suppress prostate cancer. Here, we show that auriculasin, derived from Flemingia philippinensis, induces significant cell death and apoptosis via ROS generation in prostate cancer cells. Auriculasin treatment resulted in selective apoptotic cell death in LNCaP prostate cancer cells, characterized by DNA fragmentation, accumulation of sub-G1 cell population, cleavage of poly (ADP-ribose) polymerase (PARP), regulation of Bax/Bcl-2 ratio, increase of cytosolic apoptosis-inducing factor (AIF) and endonuclease G (EndoG), in addition to inhibiting tumor growth in a xenograft mouse model. Interestingly, auriculasin-induced apoptosis did not result in caspase-3, -8, and -9 activations. We found that auriculasin treatment decreased phosphorylation of AKT/mTOR/p70s6k in a dose- and time-dependent manner. Further, cellular ROS levels increased in LNCaP cells treated with auriculasin and blocking ROS accumulation with ROS scavengers resulted in inhibition of auriculasin-induced PARP cleavage, AIF increase, upregulation of Bax/Bcl-2 ratio, and decrease in AKT/mTOR phosphorylation. Taken together, these data suggest that auriculasin targets ROS-mediated caspase-independent pathways and suppresses PI3K/AKT/mTOR signaling, which leads to apoptosis and decreased tumor growth.
Collapse
Affiliation(s)
- Hyun-Dong Cho
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ju-Hye Lee
- Functional Food and Nutrition Division, Department of Agro-Food Resource, National Academy of Agricultural Science, Rural Development Administration, Wanju, 55365, Republic of Korea
| | - Kwang-Deog Moon
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ki-Hun Park
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
26
|
Fann LY, Chen Y, Chu DC, Weng SJ, Chu HC, Wu ATH, Lee JF, Ali AAA, Chen TC, Huang HS, Ma KH. Identification and preclinical evaluation of the small molecule, NSC745887, for treating glioblastomas via suppressing DcR3-associated signaling pathways. Oncotarget 2017; 9:11922-11937. [PMID: 29552282 PMCID: PMC5844718 DOI: 10.18632/oncotarget.23714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/11/2017] [Indexed: 11/25/2022] Open
Abstract
The small-molecule naphtha [2,3-f]quinoxaline-7,12-dione (NSC745887) can effectively inhibit the proliferation of various cancers by trapping DNA-topoisomerase cleavage. The aim of this study was to elucidate cellular responses of NSC745887 in human glioblastoma multiforme (GBM, U118MG and U87MG cells) and investigate the underlying molecular mechanisms. NSC745887 reduced the cell survival rate and increased the sub-G1 population in dose- and time-dependent manners in GBM cells. Moreover, NSC745887 increased expression of γH2AX and caused DNA fragmentation leading to DNA damage. Furthermore, Annexin V/propidium iodide and Br-dTP staining showed the apoptotic effect of NSC745887 in GBM cells. DNA repair proteins of ataxia-telangiectasia mutated (ATM), ATM and Rad3-related, and decoy receptor 3 also decreased with NSC745887 treatment. In addition, NSC745887 caused apoptosis by the caspase-8/9-caspase-3-poly(ADP-ribose) polymerase cascade. An in vivo study indicated that NSC745887 suppressed the [18F]-FDG-specific uptake value in brain tumors. Histological staining also indicated a decrease in Ki-67 and increases in γH2AX and cleaved caspase-3 in the brain tumor area. These data provide preclinical evidence for NSC745887 as a potential new small molecule drug for managing glioblastomas.
Collapse
Affiliation(s)
- Li-Yun Fann
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ying Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Da-Chen Chu
- Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC
| | - Shao-Ju Weng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Heng-Cheng Chu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Alexander T H Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jiann-Fong Lee
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ahmed Atef Ahmed Ali
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsung-Chih Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsu-Shan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kuo-Hsing Ma
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
27
|
Chang MC, Lee JJ, Chen YJ, Lin SI, Lin LD, Jein-Wen Liou E, Huang WL, Chan CP, Huang CC, Jeng JH. Lysophosphatidylcholine induces cytotoxicity/apoptosis and IL-8 production of human endothelial cells: Related mechanisms. Oncotarget 2017; 8:106177-106189. [PMID: 29290940 PMCID: PMC5739725 DOI: 10.18632/oncotarget.22425] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/29/2017] [Indexed: 12/31/2022] Open
Abstract
Increased levels of oxidized low-density lipoprotein oxLDL) are shown to elevate the risk of cardiovascular diseases such as atherosclerosis, thrombosis, stroke, and myocardial infarction. This is possibly due to the toxic effects of oxLDLs on vascular cells. Various oxLDLs including lysophosphatidylcholine (LPC) and 7-ketocholesterol injure vascular endothelial cells and stimulate inflammatory reaction. However the toxicity of LPC on endothelial cells is not clear. In this study, human endothelial cells were exposed to LPC. Cytotoxicity was measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Propidium iodide (PI) staining or PI/Annexin V dual staining flow cytometry were used to determine cell cycle progression and apoptosis. Reactive oxygen species (ROS) level was analyzed by DCFH-DA labeling flow cytometry. RNA and protein expression of endothelial cells was studied by reverse transcriptase-polymerase chain reaction and western blotting. IL-8 secretion was measured by enzyme-linked immunosorbant assay. LPC showed cytotoxicity to endothelial cells (>50 µg/ml). LPC induced cell cycle arrest and apoptosis with concomitant inhibition of cdc2 and cyclin B1 expression. LPC stimulated intracellular ROS production and ATM/Chk2, ATR/Chk1 and Akt activation. IL-8 expression and secretion in endothelial cells were induced by LPC. LPC-induced apoptosis, and IL-8 expression/secretion was attenuated by LY294002, a PI3K/Akt inhibitor. These results reveal that LPC is involved in the pathogenesis of atherosclerosis and vascular diseases by stimulation of inflammation and injury to endothelial cells. These events are related to ROS, ATM/Chk2, ATR/Chk2 and PI3K/Akt signaling. Understanding the toxic mechanisms of LPC is useful for future prevention and treatment atherosclerosis.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Biomedical Science Team, Chang Gung University of Science and Technology, Kwei-Shan, Taoyuan City, Taiwan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Jang-Jaer Lee
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Jane Chen
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-I Lin
- Department of Dentistry, Municipal Taoyuan Hospital, Taoyuan City, Taiwan
| | - Li-Deh Lin
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | | | - Wei-Ling Huang
- Department of Dentistry, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chiu-Po Chan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chi-Chia Huang
- Department of Dentistry, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
28
|
Zhang YH, Huang T, Chen L, Xu Y, Hu Y, Hu LD, Cai Y, Kong X. Identifying and analyzing different cancer subtypes using RNA-seq data of blood platelets. Oncotarget 2017; 8:87494-87511. [PMID: 29152097 PMCID: PMC5675649 DOI: 10.18632/oncotarget.20903] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022] Open
Abstract
Detection and diagnosis of cancer are especially important for early prevention and effective treatments. Traditional methods of cancer detection are usually time-consuming and expensive. Liquid biopsy, a newly proposed noninvasive detection approach, can promote the accuracy and decrease the cost of detection according to a personalized expression profile. However, few studies have been performed to analyze this type of data, which can promote more effective methods for detection of different cancer subtypes. In this study, we applied some reliable machine learning algorithms to analyze data retrieved from patients who had one of six cancer subtypes (breast cancer, colorectal cancer, glioblastoma, hepatobiliary cancer, lung cancer and pancreatic cancer) as well as healthy persons. Quantitative gene expression profiles were used to encode each sample. Then, they were analyzed by the maximum relevance minimum redundancy method. Two feature lists were obtained in which genes were ranked rigorously. The incremental feature selection method was applied to the mRMR feature list to extract the optimal feature subset, which can be used in the support vector machine algorithm to determine the best performance for the detection of cancer subtypes and healthy controls. The ten-fold cross-validation for the constructed optimal classification model yielded an overall accuracy of 0.751. On the other hand, we extracted the top eighteen features (genes), including TTN, RHOH, RPS20, TRBC2, in another feature list, the MaxRel feature list, and performed a detailed analysis of them. The results indicated that these genes could be important biomarkers for discriminating different cancer subtypes and healthy controls.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China.,Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yu Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lan-Dian Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yudong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| |
Collapse
|
29
|
Wang M, Liu G, Shan GP, Wang BB. In Vivo and In Vitro Effects of ATM/ATR Signaling Pathway on Proliferation, Apoptosis, and Radiosensitivity of Nasopharyngeal Carcinoma Cells. Cancer Biother Radiopharm 2017; 32:193-203. [PMID: 28820634 DOI: 10.1089/cbr.2017.2212] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Ming Wang
- Department of Otolaryngology Head and Neck Surgery, Tianjin Huanhu Hospital, Tianjin, People's Republic of China
| | - Gang Liu
- Department of Otolaryngology Head and Neck Surgery, Tianjin Huanhu Hospital, Tianjin, People's Republic of China
| | - Guo-Ping Shan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, People's Republic of China
| | - Bing-Bing Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, People's Republic of China
| |
Collapse
|
30
|
Hu A, Huang JJ, Zhang JF, Dai WJ, Li RL, Lu ZY, Duan JL, Li JP, Chen XP, Fan JP, Xu WH, Zheng HL. Curcumin induces G2/M cell cycle arrest and apoptosis of head and neck squamous cell carcinoma in vitro and in vivo through ATM/Chk2/p53-dependent pathway. Oncotarget 2017; 8:50747-50760. [PMID: 28881600 PMCID: PMC5584201 DOI: 10.18632/oncotarget.17096] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/31/2017] [Indexed: 12/22/2022] Open
Abstract
Studies have demonstrated that curcumin (CUR) exerts its tumor suppressor function in a variety of human cancers including head and neck squamous cell carcinoma (HNSCC). However, the exact underlying molecular mechanisms remain obscure. Here, we aim to test whether CUR affects ATM/Chk2/p53 signaling pathway, leading to the induction of cell cycle arrest, inhibition of angiogenesis of HNSCC in vitro and in vivo. To this end, we conducted multiple methods such as MTT assay, Invasion assay, Flow cytometry, Western blotting, RT-PCR, and transfection to explore the functions and molecular insights of CUR in HNSCC. We observed that CUR significantly induced apoptosis and cell cycle arrest, inhibited angiogenesis in HNSCC. Mechanistically, we demonstrated that CUR markedly up-regulated ATM expression and subsequently down-regulated HIF-1α expression. Blockage of ATM production totally reversed CUR induced cell cycle arrest as well as anti-angiogenesis in HNSCC. Moreover, our results demonstrated that CUR exerts its antitumor activity through targeting ATM/Chk2/p53 signal pathway. In addition, the results of xenograft experiments in mice were highly consistent with in vitro studies. Collectively, our findings suggest that targeting ATM/Chk2/p53 signal pathway by CUR could be a promising therapeutic approach for HNSCC prevention and therapy.
Collapse
Affiliation(s)
- An Hu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Jing-Juan Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Jing-Fei Zhang
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Wei-Jun Dai
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Rui-Lin Li
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Zhao-Yang Lu
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jun-Li Duan
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Ji-Ping Li
- Department of Otolaryngology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Pudong New Area, Shanghai, 200127, China
| | - Xiao-Ping Chen
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Jing-Ping Fan
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China.,Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Wei-Hua Xu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Hong-Liang Zheng
- Department of Otolaryngology-Head and Neck Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
31
|
Liu R, Tang J, Ding C, Liang W, Zhang L, Chen T, Xiong Y, Dai X, Li W, Xu Y, Hu J, Lu L, Liao W, Lu X. The depletion of ATM inhibits colon cancer proliferation and migration via B56γ2-mediated Chk1/p53/CD44 cascades. Cancer Lett 2017; 390:48-57. [PMID: 28093285 DOI: 10.1016/j.canlet.2016.12.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/15/2016] [Accepted: 12/24/2016] [Indexed: 02/07/2023]
Abstract
Ataxia-telangiectasia mutated (ATM) protein kinase is a major guardian of genomic stability, and its well-established function in cancer is tumor suppression. Here, we report an oncogenic role of ATM. Using two isogenic sets of human colon cancer cell lines that differed only in their ATM status, we demonstrated that ATM deficiency significantly inhibits cancer cell proliferation, migration, and invasion. The tumor-suppressive function of ATM depletion is not modulated by the compensatory activation of ATR, but it is associated with B56γ2-mediated Chk1/p53/CD44 signaling pathways. Under normal growth conditions, the depletion of ATM prevents B56γ2 ubiquitination and degradation, which activates PP2A-mediated Chk1/p53/p21 signaling pathways, leading to senescence and cell cycle arrest. CD44 was validated as a novel ATM target based on its ability to rescue cell migration and invasion defects in ATM-depleted cells. The activation of p53 induced by ATM depletion suppresses CD44 transcription, thus resulting in epithelial-mesenchymal transition (EMT) and cell migration suppression. Our study suggests that ATM has tumorigenic potential in post-formed colon neoplasia, and it supports ATM as an appealing target for improving cancer therapy.
Collapse
Affiliation(s)
- Rui Liu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiajia Tang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Chaodong Ding
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Weicheng Liang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Li Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Tianke Chen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Xiong
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaowei Dai
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenfeng Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yunsheng Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jin Hu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Liting Lu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wanqin Liao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xincheng Lu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
32
|
Choi YJ, Choi YK, Lee KM, Cho SG, Kang SY, Ko SG. SH003 induces apoptosis of DU145 prostate cancer cells by inhibiting ERK-involved pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:507. [PMID: 27927199 PMCID: PMC5142381 DOI: 10.1186/s12906-016-1490-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/17/2016] [Indexed: 01/22/2023]
Abstract
Background Herbal medicines have been used in cancer treatment, with many exhibiting favorable side effect and toxicity profiles compared with conventional chemotherapeutic agents. SH003 is a novel extract from Astragalus membranaceus, Angelica gigas, and Trichosanthes Kirilowii Maximowicz combined at a 1:1:1 ratio that impairs the growth of breast cancer cells. This study investigates anti-cancer effects of SH003 in prostate cancer cells. Methods SH003 extract in 30% ethanol was used to treat the prostate cancer cell lines DU145, LNCaP, and PC-3. Cell viability was determined by MTT and BrdU incorporation assays. Next, apoptotic cell death was determined by Annexin V and 7-AAD double staining methods. Western blotting was conducted to measure protein expression levels of components of cell death and signaling pathways. Intracellular reactive oxygen species (ROS) levels were measured using H2DCF-DA. Plasmid-mediated ERK2 overexpression in DU145 cells was used to examine the effect of rescuing ERK2 function. Results were analyzed using the Student’s t-test and P-values < 0.05 were considered to indicate statistically-significant differences. Results Our data demonstrate that SH003 induced apoptosis in DU145 prostate cancer cells by inhibiting ERK signaling. SH003 induced apoptosis of prostate cancer cells in dose-dependent manner, which was independent of androgen dependency. SH003 also increased intracellular ROS levels but this is not associated with its pro-apoptotic effects. SH003 inhibited phosphorylation of Ras/Raf1/MEK/ERK/p90RSK in androgen-independent DU145 cells, but not androgen-dependent LNCaP and PC-3 cells. Moreover, ERK2 overexpression rescued SH003-induced apoptosis in DU145 cells. Conclusions SH003 induces apoptotic cell death of DU145 prostate cancer cells by inhibiting ERK2-mediated signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1490-5) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Li S, Wu L, Feng J, Li J, Liu T, Zhang R, Xu S, Cheng K, Zhou Y, Zhou S, Kong R, Chen K, Wang F, Xia Y, Lu J, Zhou Y, Dai W, Guo C. In vitro and in vivo study of epigallocatechin-3-gallate-induced apoptosis in aerobic glycolytic hepatocellular carcinoma cells involving inhibition of phosphofructokinase activity. Sci Rep 2016; 6:28479. [PMID: 27349173 PMCID: PMC4923908 DOI: 10.1038/srep28479] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022] Open
Abstract
Glycolysis, as an altered cancer cell-intrinsic metabolism, is an essential hallmark of cancer. Phosphofructokinase (PFK) is a metabolic sensor in the glycolytic pathway, and restricting the substrate availability for this enzyme has been researched extensively as a target for chemotherapy. In the present study, we investigated that the effects of epigallocatechin-3-gallate (EGCG), an active component of green tea, on inhibiting cell growth and inducing apoptosis by promoting a metabolic shift away from glycolysis in aerobic glycolytic hepatocellular carcinoma (HCC) cells. EGCG modulated the oligomeric structure of PFK, potentially leading to metabolic stress associated apoptosis and suggesting that EGCG acts by directly suppressing PFK activity. A PFK activity inhibitor enhanced the effect, while the allosteric activator reversed EGCG-induced HCC cell death. PFK siRNA knockdown-induced apoptosis was not reversed by the activator. EGCG enhanced the effect of sorafenib on cell growth inhibition in both aerobic glycolytic HCC cells and in a xenograft mouse model. The present study suggests a potential role for EGCG as an adjuvant in cancer therapy, which merits further investigation at the clinical level.
Collapse
Affiliation(s)
- Sainan Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rong Zhang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The Shanghai Tenth Hospital School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The Shanghai Tenth Hospital School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Keran Cheng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Yuqing Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Shunfeng Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Rui Kong
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|