1
|
Varlı M, Ji M, Kim E, Kim SJ, Choi B, Ha HH, Kim KK, Paik MJ, Kim H. Emodin disrupts the KITENIN oncogenic complex by binding ErbB4 and suppresses colorectal cancer progression in dual blockade with KSRP-binding compound. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156247. [PMID: 39586126 DOI: 10.1016/j.phymed.2024.156247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The KITENIN/ErbB4 complex has been reported to participate in metastasis, which is the principal reason of death in most colorectal cancer patients. PURPOSE New therapeutics need to be developed to suppress the malignant effects of the KITENIN/ErbB4 complex, which is related to drug resistance. The present study aimed to evaluate changes in cancer cell invasion capacity, transcriptional regulators, and cellular bioenergetics after targeting the KITENIN/ErbB4 complex with emodin. Moreover, we aimed to reveal the mechanistic effects of emodin and observe the dual blockade effects of ErbB4-targeted therapy with KH-type splicing regulatory protein (KSRP) and search for new alternative blockade pathways. METHODS Using in vitro, in vivo, molecular-docking, and metabolomics studies, we evaluated the anticancer effect of emodin alone or in combination with DKCC14S. RESULTS Emodin treatment decreased KITENIN and ErbB4 protein levels. The dysfunctional KITENIN/ErbB4 complex suppressed KITENIN-mediated cell invasion and downregulated AP-1 activity, aerobic glycolysis, and the levels of transcriptional regulators associated with cell metabolism. We conclude that emodin targets the KITENIN/ErbB4 complex and offering a novel mechanism by which it disrupts KITENIN-mediated signaling. Furthermore, we were demonstrated that the dual blocking effect of emodin and DKC-C14S on the KITENIN complex showed synergistic effects in suppressing colorectal cancer progression under in cell-based and animal assay. CONCLUSION The results suggest that co-treatment with ErbB4 and KSRP-binding compounds could constitute a potential strategy for controlling colorectal cancer progression by disrupting the KITENIN complex.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Moongi Ji
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Eunae Kim
- College of Pharmacy, Chosun University, 146 Chosundae-gil, Gwangju 61452, Republic of Korea.
| | - Sung Jin Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea.
| | - Byeongchan Choi
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Hyung-Ho Ha
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea.
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea.
| |
Collapse
|
2
|
Ye B, Cai X, Liang X, Chen Y, Dai S, Huang Z, Huang W, Zhang L, Wang Z, Xing J, Lai X, Huang Z, Jia Z. Emodin Suppresses NLRP3/GSDMD-induced Inflammation via the TLR4/MyD88/NF-κB Signaling Pathway in Atherosclerosis. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07659-w. [PMID: 39715879 DOI: 10.1007/s10557-024-07659-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/25/2024]
Abstract
PURPOSE Inflammatory responses induced by NLRP3 inflammasome contribute to the progression of atherosclerosis. This study seeks to investigate the effect of emodin on the NLRP3 inflammasome in atherogenesis and to probe the underlying mechanism. METHODS ApoE-knockout (ApoE-/-) mice were treated with a high-fat diet (HFD) for 12 weeks and intragastrically with emodin for 6 weeks. Human mononuclear cell line THP-1 was pretreated with emodin or signaling pathway inhibitors and induced into macrophages using phorbol 12-myristate 13-acetate (PMA) for 48 h. The NLRP3-mediated inflammatory response was studied both in vivo and in vitro. The level of the inflammation was detected by western blot, real-time PCR analysis, and ELISA. RESULTS Emodin attenuated atherosclerotic lesions in HFD-treated ApoE-/- mice. Emodin dramatically decreased the expression of NLRP3, GSDMD, IL-1β, and IL-18 in HFD-treated ApoE-/- mice and PMA-induced macrophages. Moreover, emodin significantly hindered the activation of nuclear factor kappa-B (NF-κB) by inhibiting the formation of the TLR4/MyD88 complex in PMA-induced macrophages. CONCLUSION Our data demonstrate that emodin can inhibit the development of atherosclerotic plaques by alleviating NLRP3/GSDMD-induced inflammation through repressing the TLR4/MyD88/NF-κB signaling pathway in macrophages. This finding suggests that emodin can be a potential candidate for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Cardiology, Panvascular Disease Management Center (PDMC), Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, WenZhou, ZheJiang, China
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Xueli Cai
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Xiaohe Liang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Yunxuan Chen
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Shanshan Dai
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, WenZhou, Zhejiang, China
| | - Zhuqi Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Weijian Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Lei Zhang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Zixuan Wang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Jincheng Xing
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China
| | - Xianhui Lai
- Department of Cardiology, Yuhuan County People's Hospital of Zhejiang Province, Taizhou, China.
| | - Zhouqing Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, ZheJiang, China.
| | - Zhuyin Jia
- Department of Cardiology, Panvascular Disease Management Center (PDMC), Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, WenZhou, ZheJiang, China.
| |
Collapse
|
3
|
Inclusion complexation of emodin with various β-cyclodextrin derivatives: Preparation, characterization, molecular docking, and anticancer activity. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
4
|
Zinc Supplementation Enhances the Pro-Death Function of UPR in Lymphoma Cells Exposed to Radiation. BIOLOGY 2022; 11:biology11010132. [PMID: 35053130 PMCID: PMC8773084 DOI: 10.3390/biology11010132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 02/05/2023]
Abstract
Simple Summary It is of fundamental importance to find strategies able to reduce the minimum doses of anticancer treatments, such as radiations, and concomitantly maintain efficient killing of cancer cells. The interconnection between ER stress and DNA repair may represent a promising approach to obtaining this goal. Here we found that pretreatment of lymphoma cells with Zinc chloride rendered these cells more sensitive to 2 Gy radiation. The exacerbation of ER stress and the activation pro-death function of UPR were among the underlying mechanisms leading to higher cytotoxicity of Zinc/radiation combination treatment. This evidence encourages the use of Zinc to reduce the doses of radiation in the treatment of lymphoma cells, allowing a high cytotoxicity to be obtained while minimizing the side effects. Abstract We have previously shown that Zinc supplementation triggered ER stress/UPR in cancer cells undergoing treatment by genotoxic agents, reactivated wtp53 in cancer cells harboring mutant p53 (mutp53) and potentiated the activity of wtp53 in those carrying wtp53. In this study, we used Zinc chloride alone or in combination with 2 Gy radiation to treat Primary Effusion Lymphoma (PEL) cells, an aggressive B-cell lymphoma associated with KSHV that harbors wt or partially functioning p53. We found that Zinc triggered a mild ER stress/UPR in these lymphoma cells and activated ERK1/2, molecule known to sustain cell survival in the course of UPR activation. In combination with radiations, Zinc triggered a stronger p53 activation that counteracted its mediated ERK1/2 phosphorylation, further upregulating the UPR molecule CHOP and promoting cell death. These data suggest that Zinc supplementation could be a promising strategy to reduce the doses of radiation and possibly of other DNA-damaging agents to obtain an efficient capacity to induce lymphoma cell death.
Collapse
|
5
|
Is Emodin with Anticancer Effects Completely Innocent? Two Sides of the Coin. Cancers (Basel) 2021; 13:cancers13112733. [PMID: 34073059 PMCID: PMC8198870 DOI: 10.3390/cancers13112733] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Many anticancer active compounds are known to have the capacity to destroy pathologically proliferating cancer cells in the body, as well as to destroy rapidly proliferating normal cells. Despite remarkable advances in cancer research over the past few decades, the inclusion of natural compounds in researches as potential drug candidates is becoming increasingly important. However, the perception that the natural is reliable is an issue that needs to be clarified. Among the various chemical classes of natural products, anthraquinones have many biological activities and have also been proven to exhibit a unique anticancer activity. Emodin, an anthraquinone derivative, is a natural compound found in the roots and rhizomes of many plants. The anticancer property of emodin, a broad-spectrum inhibitory agent of cancer cells, has been detailed in many biological pathways. In cancer cells, these molecular mechanisms consist of suppressing cell growth and proliferation through the attenuation of oncogenic growth signaling, such as protein kinase B (AKT), mitogen-activated protein kinase (MAPK), HER-2 tyrosine kinase, Wnt/-catenin, and phosphatidylinositol 3-kinase (PI3K). However, it is known that emodin, which shows toxicity to cancer cells, may cause kidney toxicity, hepatotoxicity, and reproductive toxicity especially at high doses and long-term use. At the same time, studies of emodin, which has poor oral bioavailability, to transform this disadvantage into an advantage with nano-carrier systems reveal that natural compounds are not always directly usable compounds. Consequently, this review aimed to shed light on the anti-proliferative and anti-carcinogenic properties of emodin, as well as its potential toxicities and the advantages of drug delivery systems on bioavailability.
Collapse
|
6
|
Nguyen THP, Kumar VB, Ponnusamy VK, Mai TTT, Nhat PT, Brindhadevi K, Pugazhendhi A. Phytochemicals intended for anticancer effects at preclinical levels to clinical practice: Assessment of formulations at nanoscale for non-small cell lung cancer (NSCLC) therapy. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
7
|
Ulus G. Antiangiogenic properties of lichen secondary metabolites. Phytother Res 2021; 35:3046-3058. [PMID: 33587324 DOI: 10.1002/ptr.7023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 12/29/2020] [Accepted: 01/03/2021] [Indexed: 12/28/2022]
Abstract
Lichens are symbiotic organisms which are composed fungi and algae and/or cyanobacteria. They produce a variety of characteristic secondary metabolites. Such substances have various biological properties including antimicrobial, antiviral, and antitumor activities. Angiogenesis, the growth of new vessels from pre-existing vessels, contributes to numerous diseases including cancer, arthritis, atherosclerosis, infectious, and immune disorders. Antiangiogenic therapy is a promising approach for the treatment of such diseases by inhibiting the new vessel formation. Technological advances have led to the development of various antiangiogenic agents and have made possible antiangiogenic therapy in many diseases associated with angiogenesis. Some lichens and their metabolites are used in the drug industry, but many have not yet been tested for their antiangiogenic effects. The cytotoxic and angiogenic capacities of lichen-derived small molecules have been demonstrated in vivo and in vitro experiments. Therefore, some of them may be used as antiangiogenic agents in the future. The secondary compounds of lichen whose antiangiogenic effect has been studied in the literature are usnic acid, barbatolic acid, vulpinic acid, olivetoric acid, emodin, secalonic acid D, and parietin. In this article, we review the antiangiogenic effects and cellular targets of these lichen-derived metabolites.
Collapse
Affiliation(s)
- Gönül Ulus
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| |
Collapse
|
8
|
Zhang N, Wang J, Sheng A, Huang S, Tang Y, Ma S, Hong G. Emodin Inhibits the Proliferation of MCF-7 Human Breast Cancer Cells Through Activation of Aryl Hydrocarbon Receptor (AhR). Front Pharmacol 2021; 11:622046. [PMID: 33542691 PMCID: PMC7850984 DOI: 10.3389/fphar.2020.622046] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
Natural products have proved to be a promising source for the development of potential anticancer drugs. Emodin, a natural compound from Rheum palmatum, is used to treat several types of cancers, including lung, liver, and pancreatic. However, there are few reports regarding its use in the treatment of breast cancer. Thus, the therapeutic effect and mechanism of emodin on MCF-7 human breast cancer cells were investigated in this study. Morphological observations and cell viability were evaluated to determine the anti-proliferation activity of emodin. Network pharmacology and molecular docking were performed to screen the potential targets. Western blot analysis was used to explore a potential antitumor mechanism. The results showed that emodin (50–100 μmol/L) could significantly inhibit the proliferation of MCF-7 cells in a time and dose-dependent manner. Furthermore, virtual screening studies indicated that emodin was a potent aryl hydrocarbon receptor (AhR) agonist in chemotherapy for breast cancer. Finally, when MCF-7 cells were treated with emodin (100 μmol/L) for 24 h, the AhR and cytochrome P450 1A1 (CYP1A1) protein expression levels were significantly upregulated compared with the control group. Our study indicated that emodin exhibited promising antitumor activity in MCF-7 cells, likely through activation of the AhR-CYP1A1 signaling pathway. These findings lay a foundation for the application of emodin in breast cancer treatment.
Collapse
Affiliation(s)
- Ning Zhang
- Life and Health College, Anhui Science and Technology University, Fengyang, China.,School of Chemical Engineering, Anhui University of Science and Technology, Huainan, China.,Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Jiawen Wang
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Aimin Sheng
- School of Chemical Engineering, Anhui University of Science and Technology, Huainan, China
| | - Shuo Huang
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin Hospital, Tianjin, China
| | - Yanyan Tang
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin Hospital, Tianjin, China
| | - Shitang Ma
- Life and Health College, Anhui Science and Technology University, Fengyang, China
| | - Ge Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| |
Collapse
|
9
|
Verebová V, Beneš J, Staničová J. Biophysical Characterization and Anticancer Activities of Photosensitive Phytoanthraquinones Represented by Hypericin and Its Model Compounds. Molecules 2020; 25:E5666. [PMID: 33271809 PMCID: PMC7731333 DOI: 10.3390/molecules25235666] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Photosensitive compounds found in herbs have been reported in recent years as having a variety of interesting medicinal and biological activities. In this review, we focus on photosensitizers such as hypericin and its model compounds emodin, quinizarin, and danthron, which have antiviral, antifungal, antineoplastic, and antitumor effects. They can be utilized as potential agents in photodynamic therapy, especially in photodynamic therapy (PDT) for cancer. We aimed to give a comprehensive summary of the physical and chemical properties of these interesting molecules, emphasizing their mechanism of action in relation to their different interactions with biomacromolecules, specifically with DNA.
Collapse
Affiliation(s)
- Valéria Verebová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
| | - Jiří Beneš
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| | - Jana Staničová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| |
Collapse
|
10
|
Guo H, Liu F, Yang S, Xue T. Emodin alleviates gemcitabine resistance in pancreatic cancer by inhibiting MDR1/P-glycoprotein and MRPs expression. Oncol Lett 2020; 20:167. [PMID: 32934734 PMCID: PMC7471752 DOI: 10.3892/ol.2020.12030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Gemcitabine is a gold standard chemotherapeutic agent for pancreatic cancer. However, gemcitabine has limited effectiveness due to the short-term development of chemoresistance. Emodin, a natural anthraquinone derivative isolated from the roots of rheumatic palm leaves prevents immunosuppression and exerts anticancer effects. The present study aimed to evaluate the effect of emodin on gemcitabine resistance. Gemcitabine-resistant PANC-1 pancreatic cancer cell xenografts were established in athymic mice, which were randomly assigned into four treatments groups as follows: Gemcitabine group, Emodin group, Gemcitabine+Emodin group and Negative control group. Body weight, tumor volume and tumor weight were measured over the course of treatment. The effect of each treatment on tumor tissue proliferation and apoptosis from nude mice was evaluated by using immunohistochemistry. The effect of each treatment on the proliferation of gemcitabine-resistant PANC-1 cells was also determined by using the Cell Counting Kit-8. Then, reverse transcription-quantitative (RT-q) PCR and western blotting were used to detect the mRNA and protein expression, respectively, of multidrug resistance gene 1 (MDR1) and the drug resistance-related proteins MRP1 and MRP5. The function and expression level of DR1 gene product, p-glycoprotein, was also analysed by flow cytometry and RT-qPCR, respectively. The results demonstrated that the combination of gemcitabine and emodin significantly reduced xenograft volume and reduced tumor growth in mice compared with treatment with gemcitabine or emodin only. In addition, emodin treatment reduced resistance to gemcitabine, which was characterized by the downregulation of P-glycoprotein, MRP1 and MRP5 expression in the group receiving combination treatment. The level of P-glycoprotein was also decreased in the group treated with gemcitabine+emodin compared with the single treatment groups. Taken together, these results demonstrated that emodin enhanced gemcitabine efficacy in tumor treatment and alleviated gemcitabine resistance in PANC-1 cell xenografts in mice via suppressing MDR1/P-glycoprotein and MRP expression.
Collapse
Affiliation(s)
- Hongchun Guo
- Department of General Surgery, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Feng Liu
- Department of General Surgery, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Shuguang Yang
- Department of Neurosurgery, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Tao Xue
- Department of Trauma Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
11
|
Jänicke P, Lennicke C, Meister A, Seliger B, Wessjohann LA, Kaluđerović GN. Fluorescent spherical mesoporous silica nanoparticles loaded with emodin: Synthesis, cellular uptake and anticancer activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111619. [PMID: 33321661 DOI: 10.1016/j.msec.2020.111619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
The natural product emodin (EO) exhibits anti-inflammatory, antiangiogenesis and antineoplastic properties in vitro and in vivo. Due to its biological properties as well as its fluorescence, EO can be useful in pharmacology and pharmacokinetics. To enhance its selectivity to cancer cells, EO was loaded into non-fluorescent and novel fluorescent spherical mesoporous nanoparticles bearing N-methyl isatoic anhydride (SNM~M) or lissamine rhodamine B sulfonyl moieties (SNM~L). The propylamine functionalized mesoporous silica nanomaterial (SNM) were characterized by powder X-ray diffraction (XRD), nitrogen gas sorption, scanning electron microscopy (SEM), transmission electron microscopy (TEM), fluorescence spectroscopy, thermogravimetric analysis (TGA) and UV spectroscopy. The cytotoxicity of EO-loaded nanoparticles was tested against the human colon carcinoma cell line HT-29. Non-loaded SNM did not affect cell proliferation, whereas those loaded with EO were at least as efficient as EO alone. It could be shown by fluorescence microscopy that the uptake of silica nanomaterial by the tumor cells occurred within 2 h and the release of EO occurred within 48 h of treatment. Flow cytometry and Western blot analysis showed that SNM containing EO induced apoptosis in HT-29 cells.
Collapse
Affiliation(s)
- Paul Jänicke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany
| | - Claudia Lennicke
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, D 06112 Halle (Saale), Germany
| | - Annette Meister
- Institute for Chemistry - Physical and Theoretical Chemistry, Martin Luther University Halle-Wittenberg, D 06099 Halle (Saale), Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, D 06112 Halle (Saale), Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany
| | - Goran N Kaluđerović
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany; Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Straße 2, 06217 Merseburg, Germany.
| |
Collapse
|
12
|
Quinonoids: Therapeutic Potential for Lung Cancer Treatment. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2460565. [PMID: 32337232 PMCID: PMC7166295 DOI: 10.1155/2020/2460565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/30/2020] [Indexed: 12/22/2022]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Owing to its high incidence and mortality, the development and discovery of novel anticancer drugs is of great importance. In recent years, many breakthroughs have been achieved in the search for effective anticancer substances from natural products. Many anticancer drugs used clinically and proven to be effective are derived from natural products. Quinonoids, including naphthoquinones, phenanthrenequinones, benzoquinones, and anthraquinones, constitute a large group of natural bioactive compounds that widely exist in higher and lower plant species. Given that most of these compounds possess anticancer effects, they are applied in many cancer studies, especially in lung cancer research. They can promote apoptosis, induce autophagy, and inhibit proliferation, angiogenesis, and cell invasion and migration. Some drugs can enhance anticancer effects when combined with other drugs. Thus, quinonoids have broad application prospects in the treatment of lung cancer. Here, we summarize the previous studies on the antilung cancer activities of quinonoids together with their underlying mechanisms and analyze the common research targets with different effects so as to provide references for the discovery of quinonoids against lung cancer.
Collapse
|
13
|
Tang W, Hong L, Dai W, Li J, Zhu H, Lin J, Yang Q, Wang Y, Lin Z, Liu M, Xiao Y, Zhang Y, Wu X, Wang J, Chen Y, Hu H, Liu S, Wang J, Xiang L. MicroRNA‑500a‑5p inhibits colorectal cancer cell invasion and epithelial‑mesenchymal transition. Int J Oncol 2020; 56:1499-1508. [PMID: 32236592 DOI: 10.3892/ijo.2020.5015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/30/2020] [Indexed: 11/06/2022] Open
Abstract
The development of malignant tumors is a series of complex processes, the majority of which have not been elucidated. The aim of the present study was to investigate the microRNAs (miRNAs/miR) that affect the migration and invasion abilities of CRC cells. Our previous reports have revealed that miR‑500a‑5p suppressed CRC cell growth and malignant transformation. The present study demonstrated that overexpression of miR‑500a‑5p reduced the expression of vimentin, while increasing the expression of E‑cadherin. Inhibition of miR‑500a‑5p resulted in spindle‑like morphological changes and reorganization of F‑actin in CRC cells. Furthermore, miR‑500a‑5p attenuated the transforming growth factor‑β signaling pathway in EMT. Additionally, emodin inhibited the miR‑500a‑5p inhibitor and suppressed the EMT process. In animal models of metastasis using nude mice, EMT and LoVo cell metastasis was modulated by miR‑500a‑5p. Therefore, the findings of the present study demonstrated that miR‑500a‑5p is associated with a positive therapeutic outcome in terms of invasion/migration of CRC cells and mesenchymal‑like cell changes.
Collapse
Affiliation(s)
- Weimei Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Linjie Hong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Weiyu Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Huiqiong Zhu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jianjiao Lin
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Qiong Yang
- Department of Gastroenterology, The Second Affiliated Hospital University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yusi Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhizhao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Mengwei Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yizhi Xiao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yaying Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Hongsong Hu
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Li Xiang
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
14
|
Qiu N, Zhao X, Liu Q, Shen B, Liu J, Li X, An L. Inclusion complex of emodin with hydroxypropyl-β-cyclodextrin: Preparation, physicochemical and biological properties. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.111151] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
15
|
Hou B, Xu S, Xu Y, Gao Q, Zhang C, Liu L, Yang H, Jiang X, Che Y. Grb2 binds to PTEN and regulates its nuclear translocation to maintain the genomic stability in DNA damage response. Cell Death Dis 2019; 10:546. [PMID: 31320611 PMCID: PMC6639399 DOI: 10.1038/s41419-019-1762-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/03/2022]
Abstract
Growth factor receptor bound protein 2 (Grb2) is an adaptor protein critical for signal transduction and endocytosis, but its role in DNA damage response (DDR) remains unknown. Here, we report that either knockdown of Grb2 or overexpression of the mutated Grb2 promotes micronuclei formation in response to oxidative stress. Furthermore, Grb2 was demonstrated to interact with phosphatase and tensin homologue (PTEN; a tumor suppressor essential for nuclear stability), and the loss of Grb2 reduced the nuclear-localized PTEN, which was further decreased upon stimulation with hydrogen peroxide (H2O2). Overexpression of the T398A-mutated, nuclear-localized PTEN reduced micronuclei frequency in the cells deficient of functional Grb2 via rescuing the H2O2-dependent expression of Rad51, a protein essential for the homologous recombination (HR) repair process. Moreover, depletion of Grb2 markedly decreased the expression of Rad51 and its interaction with PTEN. Notably, Rad51 showed a preference to immunoprecipation with the T398A-PTEN mutant, and silencing of Rad51 alone accumulated micronuclei concurring with decreased expression of both Grb2 and PTEN. Our findings indicate that Grb2 interacts with PTEN and Rad51 to regulate genomic stability in DDR by mediating the nuclear translocation of PTEN to affect the expression of Rad51.
Collapse
Affiliation(s)
- Bolin Hou
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Shanshan Xu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Yang Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Quan Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Caining Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Ling Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Huaiyi Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xuejun Jiang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Yongsheng Che
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
16
|
Differential sensitivities of bladder cancer cell lines to resveratol are unrelated to its metabolic profile. Oncotarget 2018; 8:40289-40304. [PMID: 28178690 PMCID: PMC5522211 DOI: 10.18632/oncotarget.15041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022] Open
Abstract
Resveratrol (RV) is a natural polyphenol compound with a wide range of activities, including inhibition of human bladder cancer (HBC) cell growth. Because RV is rapidly metabolized and has poor bioavailability, it is unclear whether the antitumor activity is due to RV or its metabolites. We therefore used liquid chromatography-mass spectroscopy, qRT-PCR, immunocytochemistry and western blotting to evaluate the metabolic profile and biotransformation of RV in the T24 and EJ HBC cell lines. Both T24 and EJ cells generated the same RV metabolite, RV monosulfate (RVS), and both exhibited upregulation of the RV-associated metabolic enzyme SULT1A1 (sulfotransferase). Despite these similarities, T24 cells were more sensitive to RV than EJ cells, yet T24 cells exhibited no sensitivity to an RVS mixture (84.13% RVS). Primary rat bladder epithelial cells showed no adverse effects when exposed to a therapeutic dose (100 μM) of RV. The differences in RV sensitivity between the two HBC cell lines did not reflect differences in the RV metabolic profile or SULT1A1 expression. Because RV exhibited stronger antitumor activity and better safety than RVS, we conclude that RV has significant therapeutic potential for HBC treatment, provided individual differences are considered during clinical research and application.
Collapse
|
17
|
Abstract
Recent studies suggest that neuropilin-1 (NRP-1) promotes angiogenesis mainly via VEGF and its receptors. It promotes tumorigenesis via formation of the NRP-1/ VEGF (vascular endothelial growth factor)/VEGFR2 (vascular endothelial growth factor receptor 2) complex. In addition to VEGF and its receptors, NRP-1 also binds with other growth factors such as platelet-derived growth factor (PDGF) and platelet-derived growth factor receptor (PDGFR). PDGF plays important roles in cellular proliferation and, in particular, blood vessel formation. Moreover, recent studies show that NRP-1 promotes angiogenesis via the NRP-1-ABL pathway, but independent of VEGF-VEGFR2. RAD51 is a protein involved in the signaling pathways of NRP1-ABL and PDGF(R), the expression of which is positively associated with cell radioresistance and chemoresistance. NRP-1 activates the signaling pathways of ABL and PDGF(R) to upregulate RAD51, which induces resistance to radiotherapy and chemotherapy in cancer cells. Furthermore, NRP-1 activates the tumor microenvironment by binding with fibronectin and activating ABL, thereby promoting tumor growth. Inhibition of NRP-1 may overcome the limitations of individually inhibiting the VEGF-VEGFR2 pathway in cancer therapy and provide new ideas for cancer treatment. Therefore, we review the role of NRP-1 in VEGF-VEGFR2-independent tumorigenesis.
Collapse
Affiliation(s)
- Chenxi Hu
- Department of Radiation Oncology, Lianyungang First People's Hospital, No.182, Tongguan Road, Lianyungang City, 222002, Jiangsu Province, China
| | - Xiaodong Jiang
- Department of Radiation Oncology, Lianyungang First People's Hospital, No.182, Tongguan Road, Lianyungang City, 222002, Jiangsu Province, China.
| |
Collapse
|
18
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017. [PMID: 29156754 DOI: 10.18632/oncotarget.20482] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People's Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
19
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017. [PMID: 29156754 DOI: 10.18632/oncotarget.20482]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People's Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
20
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017; 8:85759-85771. [PMID: 29156754 PMCID: PMC5689644 DOI: 10.18632/oncotarget.20482] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/25/2017] [Indexed: 01/13/2023] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People’s Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
21
|
Emodin suppresses the nasopharyngeal carcinoma cells by targeting the chloride channels. Biomed Pharmacother 2017; 90:615-625. [DOI: 10.1016/j.biopha.2017.03.088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/24/2017] [Accepted: 03/26/2017] [Indexed: 12/27/2022] Open
|
22
|
Shadrack DM, K. Ndesendo VM. Molecular Docking and ADMET Study of Emodin Derivatives as Anticancer Inhibitors of NAT2, COX2 and TOP1 Enzymes. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/cmb.2017.71001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
|
24
|
Chen T, Chen H, Wang Y, Zhang J. In vitro and in vivo antitumour activities of puerarin 6″-O-xyloside on human lung carcinoma A549 cell line via the induction of the mitochondria-mediated apoptosis pathway. PHARMACEUTICAL BIOLOGY 2016; 54:1793-1799. [PMID: 26730946 DOI: 10.3109/13880209.2015.1127980] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/06/2015] [Accepted: 12/01/2015] [Indexed: 06/05/2023]
Abstract
Context Pueraria lobata (Leguminoseae) shows cytotoxic effects against cancer cells; however, its active components remain unclear. Objective This study investigated the antitumour activity of puerarin 6″-O-xyloside (POS) on the human lung carcinoma A549 cell line. Materials and methods The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to determine the cytotoxicity of POS (at 10, 20 and 40 μM) in vitro, and xenograft nude mice were established to evaluate the antitumour effect of POS (at 40 mg/kg/d) in vivo by 15 days intraperitoneal injection (ip). To explore its mechanism of action, flow cytometry was performed to determine the pro-apoptotic effect of POS (at 10, 20 and 40 μM). Subsequently, the expression of caspase-3, caspase-7, caspase-9, Bcl-2 and Bax in A549 cells were determined. Results POS showed significant cytotoxicity toward A549 cells (p < 0.05) by inducing apoptosis. Treatment with POS significantly upregulated the levels of caspase-3 (p < 0.01), caspase-7 (p < 0.01), caspase-9 (p < 0.01) and Bax (p < 0.01) in A549 cells, and Bcl-2 was downregulated (p < 0.01). Additionally, the in vivo animal study showed that POS significantly inhibited tumour growth in A549 cells (p < 0.01). Conclusion Our study demonstrated the POS has significant antitumour activities. The mechanisms are related to increased levels of caspase-3, caspase-7, caspase-9 and Bax, and reduced levels Bcl-2.
Collapse
Affiliation(s)
- Ti Chen
- a Department of Oncology , Changshu No. 1 People's Hospital , Changshu , PR China
| | - Hui Chen
- a Department of Oncology , Changshu No. 1 People's Hospital , Changshu , PR China
| | - Ying Wang
- a Department of Oncology , Changshu No. 1 People's Hospital , Changshu , PR China
| | - Jian Zhang
- a Department of Oncology , Changshu No. 1 People's Hospital , Changshu , PR China
| |
Collapse
|
25
|
Ma CG. Effect of emodin on apoptosis in human stomach cancer cell line SGC-7901 and possible mechanisms involved. Shijie Huaren Xiaohua Zazhi 2016; 24:2463-2469. [DOI: 10.11569/wcjd.v24.i16.2463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the effect of emodin on cell apoptosis in human stomach cancer cell line SGC-7901 and to explore the related mechanisms.
METHODS: After SGC-7901 cells were treated with different concentrations of Emodin, cell apoptosis was determined by CCK-8 assay and TUNEL assay. The expression of Bcl-2, Bax, cleaved-Caspase3, cleaved-PARP, preCaspase3, and PARP was measured by Western blot. Mitochondrial membrane potential was examined by JC-1 staining and fluorescence microscopy.
RESULTS: The apoptotic rate of SGC-7901 cells was increased by emodin in a dose-dependent manner (survival rate 100.73% ± 8.97% vs 45.27% ± 3.75%, P < 0.05). Compared with the control group, treatment with emodin (60 μmol/L) significantly increased Bax expression (1.98 ± 0.12 vs 1.00 ± 0.08, P < 0.05), Caspase3 (1.73 ± 0.13 vs 0.98 ± 0.06, P < 0.05) and PARP (2.29 ± 0.17 vs 1.01 ± 0.08, P < 0.05) activation, while decreased Bcl-2 expression (0.31 ± 0.02 vs 1.01 ± 0.06, P < 0.05), Bcl-2/Bax ratio (0.14 ± 0.01 vs 1.02 ± 0.13, P < 0.05) and mitochondrial membrane potential.
CONCLUSION: Emodin accelerates the apoptosis of SGC-7901 cells via the mitochondria-dependent pathway. Our results reveal a novel role for emodin in the treatment of gastric cancer.
Collapse
|
26
|
Antitumor Effects and Mechanism of Novel Emodin Rhamnoside Derivatives against Human Cancer Cells In Vitro. PLoS One 2015; 10:e0144781. [PMID: 26682731 PMCID: PMC4684281 DOI: 10.1371/journal.pone.0144781] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/22/2015] [Indexed: 12/31/2022] Open
Abstract
A series of novel anthracene L-rhamnopyranosides compounds were designed and synthesized and their anti-proliferative activities on cancer cell lines were investigated. We found that one derivative S-8 (EM-d-Rha) strongly inhibited cell proliferation of a panel of different human cancer cell lines including A549, HepG2, OVCAR-3, HeLa and K562 and SGC-790 cell lines, and displayed IC50 values in low micro-molar ranges, which are ten folds more effective than emodin. In addition, we found EM-d-Rha (3-(2”,3”-Di-O-acetyl-α-L-rhamnopyranosyl-(1→4)-2’,3’-di-O-acetyl-α-L-rhamnopyranosyl)-emodin) substantially induced cellular apoptosis of HepG2 and OVCAR-3 cells in the early growth stage. Furthermore, EM-d-Rha led to the decrease of mitochondrial transmembrane potential, and up-regulated the express of cells apoptosis factors in a concentration- and time-dependent manner. The results indicated the EM-d-Rha may inhibit the growth and proliferation of HepG2 cells through the pathway of apoptosis induction, and the possible molecular mechanism may due to the activation of intrinsic apoptotic signal pathway.
Collapse
|
27
|
Identification of gene markers in the development of smoking-induced lung cancer. Gene 2015; 576:451-7. [PMID: 26518718 DOI: 10.1016/j.gene.2015.10.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/13/2015] [Accepted: 10/23/2015] [Indexed: 12/28/2022]
Abstract
Lung cancer is a malignant tumor with high mortality in both women and men. To study the mechanisms of smoking-induced lung cancer, we analyzed microarray of GSE4115. GSE4115 was downloaded from Gene Expression Omnibus including 78 and 85 bronchial epithelium tissue samples separately from smokers with and without lung cancer. Limma package in R was used to screen differentially expressed genes (DEGs). Hierarchical cluster analysis for DEGs was conducted using orange software and visualized by distance map. Using DAVID software, functional and pathway enrichment analyses separately were conducted for the DEGs. And protein-protein interaction (PPI) network was constructed using Cytoscape software. Then, the pathscores of enriched pathways were calculated. Besides, functional features were screened and optimized using the recursive feature elimination (RFE) method. Additionally, the support vector machine (SVM) method was used to train model. Total 1923 DEGs were identified between the two groups. Hierarchical cluster analysis indicated that there were differences in gene level between the two groups. And SVM analysis indicated that the five features had potential diagnostic value. Importantly, MAPK1 (degree=30), SRC (degree=29), SMAD4 (degree=23), EEF1A1 (degree=21), TRAF2 (degree=21) and PLCG1 (degree=20) had higher degrees in the PPI network of the DEGs. They might be involved in smoking-induced lung cancer by interacting with each other (e.g. MAPK1-SMAD4, SMAD4-EEF1A1 and SRC-PLCG1). MAPK1, SRC, SMAD4, EEF1A1, TRAF2 and PLCG1 might be responsible for the development of smoking-induced lung cancer.
Collapse
|
28
|
Abstract
Emodin (1,3,8-trihydroxy-6-methylanthraquinone) has been widely used as a traditional medicine and was shown to possess a multitude of health-promoting properties in pre-clinical studies, but its bioavailability was low due to the extensive glucuronidation in liver and intestine, hindering the development of emodin as a feasible chemopreventive agent. In this study, piperine, as a bioenhancer, was used to enhance the bioavailability of emodin by inhibiting its glucuronidation. The pharmacokinetic profiles of emodin after oral administration of emodin (20mg/kg) alone and in combination with piperine (20mg/kg) to rats were investigated via a validated LC/MS/MS method. As the in vivo pharmacokinetic studies had indicated, the AUC and Cmax of emodin were increased significantly after piperine treatment, and the glucuronidation of emodin was markedly inhibited. Our study demonstrated that piperine significantly improved the in vivo bioavailability of emodin and the influence of piperine on the pharmacokinetics of emodin may be attributed to the inhibition of glucuronidation of emodin. Further research is needed to investigate the detailed mechanism of improved bioavailability of emodin via its combination with piperine.
Collapse
|
29
|
Bailon-Moscoso N, Romero-Benavides JC, Tinitana-Imaicela F, Ostrosky-Wegman P. Medicinal plants of Ecuador: a review of plants with anticancer potential and their chemical composition. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1335-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Chen L, Liu T, Zhou J, Wang Y, Wang X, Di W, Zhang S. Citrate synthase expression affects tumor phenotype and drug resistance in human ovarian carcinoma. PLoS One 2014; 9:e115708. [PMID: 25545012 PMCID: PMC4278743 DOI: 10.1371/journal.pone.0115708] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 11/26/2014] [Indexed: 01/02/2023] Open
Abstract
Citrate synthase (CS), one of the key enzymes in the tricarboxylic acid (TCA) cycle, catalyzes the reaction between oxaloacetic acid and acetyl coenzyme A to generate citrate. Increased CS has been observed in pancreatic cancer. In this study, we found higher CS expression in malignant ovarian tumors and ovarian cancer cell lines compared to benign ovarian tumors and normal human ovarian surface epithelium, respectively. CS knockdown by RNAi could result in the reduction of cell proliferation, and inhibition of invasion and migration of ovarian cancer cells in vitro. The drug resistance was also inhibited possibly through an excision repair cross complementing 1 (ERCC1)-dependent mechanism. Finally, upon CS knockdown we observed significant increase expression of multiple genes, including ISG15, IRF7, CASP7, and DDX58 in SKOV3 and A2780 cells by microarray analysis and real-time PCR. Taken together, these results suggested that CS might represent a potential therapeutic target for ovarian carcinoma.
Collapse
Affiliation(s)
- Lilan Chen
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
| | - Ting Liu
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
| | - Yunfei Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
| | - Xinran Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
- * E-mail: (WD); (SZ)
| | - Shu Zhang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China
- * E-mail: (WD); (SZ)
| |
Collapse
|
31
|
SUN YANG, WANG XIUFENG, ZHOU QIANMEI, LU YIYU, ZHANG HUI, CHEN QILONG, ZHAO MING, SU SHIBING. Inhibitory effect of emodin on migration, invasion and metastasis of human breast cancer MDA-MB-231 cells in vitro and in vivo. Oncol Rep 2014; 33:338-46. [DOI: 10.3892/or.2014.3585] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 10/06/2014] [Indexed: 11/05/2022] Open
|
32
|
Ma L, Li W. Emodin inhibits LOVO colorectal cancer cell proliferation via the regulation of the Bcl-2/Bax ratio and cytochrome c.. Exp Ther Med 2014; 8:1225-1228. [PMID: 25187829 PMCID: PMC4151688 DOI: 10.3892/etm.2014.1900] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 06/25/2014] [Indexed: 01/04/2023] Open
Abstract
In this study, the effect of emodin and its mechanism of action were investigated in LOVO colorectal cancer cells. Cell growth was determined using a Cell Counting kit-8 assay, and the results demonstrated that emodin significantly inhibited the growth of LOVO cells in a concentration-dependent manner. In order to investigate the anticancer mechanism of emodin, reverse transcription polymerase chain reaction assays were performed to determine the B-cell lymphoma-2 (Bcl-2)/Bcl-2-associated X protein (Bax) expression ratio in LOVO colorectal cancer cells following treatment with emodin. The results showed that emodin induced a significant increase in the Bax expression level and a marked reduction of the Bcl-2 expression level in LOVO cells. In addition, emodin was found to have an inhibitory effect on the mitochondrial membrane potential and the results from the western blot analysis revealed that cytochrome c was released from the mitochondria to the cytoplasm. In combination, these results suggest that emodin inhibits cancer cell growth via the regulation of the Bcl-2/Bax ratio and by its effect on the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Liang Ma
- Department of Anorectal Surgery, The Affiliated Traditional Chinese Medicine Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Wusheng Li
- Department of Anorectal Surgery, The Affiliated Traditional Chinese Medicine Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
33
|
The combined effect of survivin-targeted shRNA and emodin on the proliferation and invasion of ovarian cancer cells. Anticancer Drugs 2014; 24:937-44. [PMID: 23921083 DOI: 10.1097/cad.0b013e328364efe0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Survivin has been shown to be highly expressed in ovarian cancers, but not normal ovarian tissue, which makes it an attractive target for ovarian cancer treatment. Emodin is a traditional Chinese medicine that has been found to inhibit proliferation and induce apoptosis in ovarian cancer cells. Thus, in our study, we combined survivin-targeted shRNA (sur-shRNA) with emodin and tested the effects of this combination on ovarian cancer cells to identify more effective therapeutics against ovarian cancer. A sur-shRNA plasmid was constructed and transfected into the ovarian cancer cell lines SKOV3 and HO8910, and the cells were cultured for 24 h. The cells were then treated with emodin for specific time periods and assessed for viability and apoptosis using the MTT assay and flow cytometry, respectively. Cell invasion was also measured using a Matrigel invasion assay. The shRNA specific for survivin effectively reduced the expression of survivin at the mRNA and protein levels in SKOV3 and HO8910 cells. Both emodin and shRNA-mediated knockdown of survivin significantly inhibited cell proliferation, induced apoptosis, and suppressed invasion in SKOV3 and HO8910 cells (P<0.05). Moreover, the combination of the agents significantly enhanced these effects (P<0.05). We found that the combination of sur-shRNA and emodin could be effective in the treatment of ovarian cancer.
Collapse
|
34
|
Way TD, Huang JT, Chou CH, Huang CH, Yang MH, Ho CT. Emodin represses TWIST1-induced epithelial-mesenchymal transitions in head and neck squamous cell carcinoma cells by inhibiting the β-catenin and Akt pathways. Eur J Cancer 2013; 50:366-78. [PMID: 24157255 DOI: 10.1016/j.ejca.2013.09.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 06/20/2013] [Accepted: 09/17/2013] [Indexed: 12/11/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the leading causes of cancer deaths worldwide. In recent studies, a crucial link has been discovered between the acquisition of metastatic traits and tumour-initiating abilities in cancer cells during the epithelial-mesenchymal transition (EMT). Herein, we demonstrated that the ectopic expression of TWIST1, the EMT regulator, in HNSCC FaDu cells triggered EMT and resulted in the acquisition of a mesenchymal phenotype. Moreover, FaDu-pFLAG-TWIST1 cancer cell populations that were induced to EMT displayed an increased proportion of cells with the CD44 marker, which is associated with tumour initiation. Interestingly, we found that emodin treatment reduced the tumour-initiating abilities and inhibited cell migration and invasion in FaDu-pFLAG-TWIST1 cells. Emodin directly inhibited TWIST1 expression, upregulated E-cadherin mRNA and protein expression, and downregulated vimentin mRNA and protein expression. Moreover, we found that emodin inhibited TWIST1 binding to the E-cadherin promoter and repressed E-cadherin transcription activity. We also found that emodin inhibited TWIST1-induced EMT by inhibiting the β-catenin and Akt pathways. More interestingly, emodin significantly inhibited TWIST1-induced invasion in vivo. Therefore, emodin might be applicable to anticancer therapy and could be a potential new therapeutic drug for HNSCC.
Collapse
Affiliation(s)
- Tzong-Der Way
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| | - Jhen-Ting Huang
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Chun-Hung Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Hung Huang
- Taiwan Advance Biopharm, New Taipei City, Taiwan; Graduate School of Biotechnology, Hung-Kuang University, Taichung, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Hematology-Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
35
|
Wei WT, Lin SZ, Liu DL, Wang ZH. The distinct mechanisms of the antitumor activity of emodin in different types of cancer (Review). Oncol Rep 2013; 30:2555-62. [PMID: 24065213 DOI: 10.3892/or.2013.2741] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/06/2013] [Indexed: 11/05/2022] Open
Abstract
Emodin, a tyrosine kinase inhibitor, is a natural anthraquinone derivative found in the roots and rhizomes of numerous plants. The inhibitory effect of emodin on mammalian cell cycle modulation in specific oncogene-overexpressing cells has formed the basis for using this compound as an anticancer drug. Previous reviews have summarized the antitumor properties of emodin. However, the specific molecular mechanisms of emodin-mediated tumor inhibition have not been completely elucidated over the last 5 years. Recently, there has been great progress in the preclinical study of the anticancer mechanisms of emodin. Our recent study revealed that emodin has therapeutic effects on pancreatic cancer through various antitumor mechanisms. Notably, the therapeutic efficacy of emodin in combination with chemotherapy was found to be higher than the comparable single chemotherapeutic regime, and the combination therapy also exhibited fewer side-effects. Despite these encouraging results, further investigation is warranted as emodin has been shown to modulate one or more key regulators of cancer growth. This review provides an overview of the distinct mechanisms of anticancer action of emodin in different body systems identified over the past 5 years. These new breakthrough findings may have important implications for targeted cancer therapy and for the future clinical use of emodin.
Collapse
Affiliation(s)
- Wei-Tian Wei
- Department of Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou 310005, P.R. China
| | | | | | | |
Collapse
|
36
|
Bae JY, Lee YS, Han SY, Jeong EJ, Lee MK, Kong JY, Lee DH, Cho KJ, Lee HS, Ahn MJ. A Comparison between Water and Ethanol Extracts of Rumex acetosa for Protective Effects on Gastric Ulcers in Mice. Biomol Ther (Seoul) 2013; 20:425-30. [PMID: 24009831 PMCID: PMC3762272 DOI: 10.4062/biomolther.2012.20.4.425] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 12/17/2022] Open
Abstract
Rumex acetosa is a perennial herb that is widely distributed across eastern Asia. Although the hot water extract of R. acetosa has been used to treat gastritis or gastric ulcers as a folk medicine, no scientific report exists for the use of this plant to treat gastric ulcers. Hence, the present study was undertaken to assess the anti-ulcer activity of water and 70% ethanol extracts obtained from R. acetosa, using an HCl/ethanol-induced gastric ulcer model in mice. Anti-inflammatory and free radical-scavenging activities of these two extracts were also evaluated and compared. As a result, the administration of R. acetosa extracts significantly reduced the occurrence of gastric ulcers. However, significant differences in protective activity against gastric ulcers were observed between the two samples. In the case of the group pretreated with an ethanol extract dosage of 100 mg/kg, the protective effect (90.9%) was higher than that of water extract (41.2%). Under histological evaluation, pretreatment with R. acetosa extracts reversed negative effects, such as inflammation, edema, moderate hemorrhaging and loss of epithelial cells, presented by HCl/ ethanol-treated stomachs. Meanwhile, R. acetosa extracts showed potent DPPH radical-scavenging activity and decreased NO production in a murine macrophage cell line, RAW 264.7, in a dose-dependent manner without affecting cellular viability. The greater anti-ulcer and NO production inhibitory activities exhibited by ethanol extracts compared to water extracts could be ascribed to the higher emodin levels, a major anthraquinone component of this plant.
Collapse
Affiliation(s)
- Ji-Yeong Bae
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 660-751, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen G, Qiu H, Ke SD, Hu SM, Yu SY, Zou SQ. Emodin regulating excision repair cross-complementation group 1 through fibroblast growth factor receptor 2 signaling. World J Gastroenterol 2013; 19:2481-2491. [PMID: 23674849 PMCID: PMC3646138 DOI: 10.3748/wjg.v19.i16.2481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 01/26/2013] [Accepted: 03/23/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the molecular mechanisms underlying the reversal effect of emodin on platinum resistance in hepatocellular carcinoma.
METHODS: After the addition of 10 μmol/L emodin to HepG2/oxaliplatin (OXA) cells, the inhibition rate (IR), 50% inhibitory concentration (IC50) and reversal index (IC50 in experimental group/IC50 in control group) were calculated. For HepG2, HepG2/OXA, HepG2/OXA/T, each cell line was divided into a control group, OXA group, OXA + fibroblast growth factor 7 (FGF7) group and OXA + emodin group, and the final concentrations of FGF7, emodin and OXA in each group were 5 ng/mL, 10 μg/mL and 10 μmol/L, respectively. Single-cell gel electrophoresis was conducted to detect DNA damage, and the fibroblast growth factor receptor 2 (FGFR2), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) and excision repair cross-complementing gene 1 (ERCC1) protein expression levels in each group were examined by Western blotting.
RESULTS: Compared with the IC50 of 120.78 μmol/L in HepG2/OXA cells, the IC50 decreased to 39.65 μmol/L after treatment with 10 μmol/L emodin; thus, the reversal index was 3.05. Compared with the control group, the tail length and Olive tail length in the OXA group, OXA + FGF7 group and OXA + emodin group were significantly increased, and the differences were statistically significant (P < 0.01). The tail length and Olive tail length were lower in the OXA + FGF7 group than in the OXA group, and this difference was also statistically significant. Compared with the OXA + FGF7 group, the tail extent, the Olive tail moment and the percentage of tail DNA were significantly increased in the OXA + emodin group, and these differences were statistically significant (P < 0.01). In comparison with its parental cell line HepG2, the HepG2/OXA cells demonstrated significantly increased FGFR2, p-ERK1/2 and ERCC1 expression levels, whereas the expression of all three molecules was significantly inhibited in HepG2/OXA/T cells, in which FGFR2 was silenced by FGFR2 shRNA. In the examined HepG2 cells, the FGFR2, p-ERK1/2 and ERCC1 expression levels demonstrated increasing trends in the OXA group and OXA + FGF7 group. Compared with the OXA group and OXA + FGF7 group, the FGFR2, p-ERK1/2, and ERCC1 expression levels were significantly lower in the OXA + emodin group, and these differences were statistically significant. In the HepG2/OXA/T cell line that was transfected with FGFR2 shRNA, the FGFR2, p-ERK1/2 and ERCC1 expression levels were significantly inhibited, but there were no significant differences in these expression levels among the OXA, OXA + FGF7 and OXA + emodin groups.
CONCLUSION: Emodin markedly reversed OXA resistance by enhancing OXA DNA damage in HepG2/OXA cells, and the molecular mechanism was related to the inhibitory effect on ERCC1 expression being mediated by the FGFR2/ERK1/2 signaling pathway.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cell Proliferation/drug effects
- DNA Damage
- DNA-Binding Proteins/metabolism
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Emodin/pharmacology
- Endonucleases/metabolism
- Fibroblast Growth Factor 7/metabolism
- Hep G2 Cells
- Humans
- Inhibitory Concentration 50
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Organoplatinum Compounds/pharmacology
- Oxaliplatin
- Phosphorylation
- RNA Interference
- Receptor, Fibroblast Growth Factor, Type 2/drug effects
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Signal Transduction/drug effects
- Transfection
Collapse
|
38
|
Chen G, Qiu H, Ke S, Hu S, Yu S, Zou S. The fibroblast growth factor receptor 2-mediated extracellular signal-regulated kinase 1/2 signaling pathway plays is important in regulating excision repair cross-complementary gene 1 expression in hepatocellular carcinoma. Biomed Rep 2013; 1:604-608. [PMID: 24648994 DOI: 10.3892/br.2013.96] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 04/15/2013] [Indexed: 12/19/2022] Open
Abstract
Excision repair cross-complementary gene 1 (ERCC1) is a downstream regulatory target of fibroblast growth factor receptor 2 (FGFR2); however, the mechanism of its action has not been elucidated. The cascades downstream of FGFR2 include the PKC, Ras/Raf/MEK/ERK, JAK/STAT and PI3K pathways. ERCC1 is considered to be a closely related downstream target gene of extracellular signal-regulated kinase (ERK)1/2, since ERCC1 mRNA and protein levels may be inhibited by the ERK inhibitor U0126. It was hypothesized that FGFR2, which specifically binds with fibroblast growth factor 7 (FGF7), may regulate ERCC1 gene expression through the ERK signaling pathway. The aim of the present study was to explore the association between the regulatory effect of FGFR2 on ERCC1 gene expression and the p-ERK1/2 signaling pathway in a drug-resistant hepatocellular carcinoma (HCC) cell line. The drug-resistant cell line HepG2/OXA and its parental cell line HepG2 were transfected with Bek shRNA in the logarithmic growth phase. Transfected and untransfected HepG2 and HepG2/OXA cells were then stimulated with FGF7 and changes in the protein expression of FGFR2, p-ERK1/2 and ERCC1 was detected with western blot analysis. Following transfection, HepG2/T and HepG2/OXA/T cells were observed to grow stably in a screening medium containing puromycin. The western blot analysis demonstrated a significant decrease in the protein expressions of FGFR2, p-ERK1/2 and ERCC1 in HepG2/T and HepG2/OXA/T cells as compared to untransfected cells. Expression of FGFR2, p-ERK1/2 and ERCC1 in HepG2/OXA cells was significantly increased compared to the parental HepG2 cells. Following stimulation with FGF7, the expression of FGFR2, p-ERK1/2 and ERCC1 was increased, with significant differences between HepG2 and HepG2/OXA cells in the expression of p-ERK1/2 and ERCC1. No differences were detected in the protein levels following Bek shRNA transfection in HepG2/T and HepG2/OXA/T cells. In conclusion, the FGFR2-mediated ERK1/2 signaling pathway in HCC cells plays an important role in the regulation of ERCC1 expression.
Collapse
Affiliation(s)
- Gang Chen
- Departments of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hong Qiu
- Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shandong Ke
- Departments of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shaoming Hu
- Departments of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shiying Yu
- Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shengquan Zou
- Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
39
|
Zhang W, Chen H, Liu DL, Li H, Luo J, Zhang JH, Li Y, Chen KJ, Tong HF, Lin SZ. Emodin sensitizes the gemcitabine-resistant cell line Bxpc-3/Gem to gemcitabine via downregulation of NF-κB and its regulated targets. Int J Oncol 2013; 42:1189-96. [PMID: 23440366 DOI: 10.3892/ijo.2013.1839] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 02/04/2013] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate whether emodin can overcome the chemoresistance of the gemcitabine-resistant cancer cell line (Bxpc-3/Gem) in vitro. The cell line Bxpc-3/Gem was derived from the human pancreatic cancer cell line Bxpc-3. We found that Bxpc-3/Gem cells were characterized by a series of morphological changes with a resistance index of 43.51 comparing with the parental cell line. Emodin reduced Bxpc-3/Gem cell proliferation in a dose-dependent manner. Emodin and gemcitabine combination treatments resulted in decreased cell proliferation and increased apoptosis in Bxpc-3/Gem cells. In addition, combination treatments resulted in downregulation of gene and protein expression of MDR-1 (P-gp), NF-κB, XIAP, survivin, as well as inhibition of NF-κB activity and P-gp function. These observations suggest that emodin may sensitize the pancreatic cancer gemcitabine-resistant cell line Bxpc-3/Gem to gemcitabine therapy via inhibition of survival signaling.
Collapse
Affiliation(s)
- Wei Zhang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
He L, Bi JJ, Guo Q, Yu Y, Ye XF. Effects of emodin extracted from Chinese herbs on proliferation of non-small cell lung cancer and underlying mechanisms. Asian Pac J Cancer Prev 2013; 13:1505-10. [PMID: 22799356 DOI: 10.7314/apjcp.2012.13.4.1505] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
To aim of this was to observe emodin-mediated cytotoxicity and its influence on Rad51 and ERCC1 expressionin non-small cell lung cancer (NSCLC). NSCLC cells were cultured in vitro with emodin at various concentrations (0, 25, 50, 75 and 100 μmol/L) for 48 h and the proliferation inhibition rate was determined by the MTT method. Then, NSCLC were treated with emodin (SK-MES-1 40 μmol/L, A549 70 μmol/L) or 20 μmol/L U0126 (an ERK inhibitor) for 48 h, or with various concentrations of emodin for 48 h and the protein and mRNA expressions of ERCC1 and Rad51 were determined by RT-PCR and Western blot assay, respectively. Emodin exerted a suppressive effect on the proliferation of NSCLC in a concentration dependent manner. Protein and mRNA expression of ERCC1 and Rad51 was also significantly decreased with the dose. Vacuolar degeneration was observed in A549 and SK-MES-1 cell lines after emodin treatment by transmission electron microscopy. Emodin may thus inhibited cell proliferation in NSCLC cells by downregulation ERCC1 and Rad51.
Collapse
Affiliation(s)
- Lin He
- Department of Pathology, Institute of Neuroscience, Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
41
|
Hsu SC, Chung JG. Anticancer potential of emodin. Biomedicine (Taipei) 2012; 2:108-116. [PMID: 32289000 PMCID: PMC7104001 DOI: 10.1016/j.biomed.2012.03.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 02/06/2012] [Accepted: 03/28/2012] [Indexed: 02/08/2023] Open
Abstract
Traditional Chinese Medicine (TCM) is widely used in clinical research due to its low toxicity, low number of side effects, and low cost. Many components of common fruits and vegetables play well-documented roles as chemopreventive or chemotherapeutic agents that suppress tumorigenesis. Anthraquinones are commonly extracted from the Polygonaceae family of plants, e.g., Rheum palmatum and Rheum officinale. Some of the major chemical components of anthraquinone and its derivatives, such as aloe-emodin, danthron, emodin, chrysophanol, physcion, and rhein, have demonstrated potential anticancer properties. This review evaluates the pharmacological effects of emodin, a major component of Aloe vera. In particular, emodin demonstrates anti-neoplastic, anti-inflammatory, anti-angiogenesis, and toxicological potential for use in pharmacology, both in vitro and in vivo. Emodin demonstrates cytotoxic effects (e.g., cell death) through the arrest of the cell cycle and the induction of apoptosis in cancer cells. The overall molecular mechanisms of emodin include cell cycle arrest, apoptosis, and the promotion of the expression of hypoxia-inducible factor 1α, glutathione S-transferase P, N-acetyltransferase, and glutathione phase I and II detoxification enzymes while inhibiting angiogenesis, invasion, migration, chemical-induced carcinogen-DNA adduct formation, HER2/neu, CKII kinase, and p34cdc2 kinase in human cancer cells. Hopefully, this summary will provide information regarding the actions of emodin in cancer cells and broaden the application potential of chemotherapy to additional cancer patients in the future.
Collapse
Affiliation(s)
- Shu-Chun Hsu
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
| |
Collapse
|
42
|
Li CL, Ma J, Zheng L, Li HJ, Li P. Determination of emodin in L-02 cells and cell culture media with liquid chromatography-mass spectrometry: application to a cellular toxicokinetic study. J Pharm Biomed Anal 2012; 71:71-8. [PMID: 22944356 DOI: 10.1016/j.jpba.2012.07.031] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/23/2012] [Accepted: 07/29/2012] [Indexed: 01/12/2023]
Abstract
The emodin-involved hepatotoxicity has been gaining increasing attention. The purpose of the present study was to evaluate the cytotoxicity of emodin on cultured human liver cells (L-02) and predict the possible relation between its cytotoxicity and cellular toxicokinetics. Cell viability and cell damage were assessed by Cell Counting Kit-8 (CCK-8) assay and phase-contrast microscopy, respectively. Cytotoxicity tests demonstrated a concentration- and time-dependent toxic effect of emodin on L-02 cells. Furthermore, emodin at concentration of 30μM led to a significant apoptosis in a time-dependent manner supported by the morphological changes of drug-treated cells. In addition, to elucidate the toxicokinetic characteristics of emodin, a highly sensitive and selective liquid chromatography-mass spectrometry (LC-MS) method was employed and validated for detecting the dynamic alteration of emodin in cells and cell culture media. The proposed method appeared to be suitable for the analysis of emodin with desirable linearity (r(2)>0.99), and satisfying precision being less than 8.7%. The range of recoveries of this method was 90.2-101.9%. The preliminary cellular toxicokinetic study revealed a time-dependent intracellular accumulation of emodin, which was consistent with its in vitro toxic effects. These findings confirmed the cytotoxicity of emodin against L-02 cells and displayed the cytotoxic manner of emodin in terms of its cellular uptake and accumulation in L-02 cells.
Collapse
Affiliation(s)
- Cui-Li Li
- State Key Laboratory of Natural Medicines (China Pharmaceutical University), No. 24 Tong jia Lane, Nanjing 210009, China
| | | | | | | | | |
Collapse
|
43
|
ERCC1 expression as a prognostic and predictive factor in patients with non-small cell lung cancer: a meta-analysis. Mol Biol Rep 2012; 39:6933-42. [PMID: 22302397 DOI: 10.1007/s11033-012-1520-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 01/24/2012] [Indexed: 02/05/2023]
Abstract
It is hypothesized that high expression of the excision repair cross-complementation group 1 (ERCC1) gene might be a positive prognostic factor, but predict decreased sensitivity to platinum-based chemotherapy. Results from the published data are inconsistent. To derive a more precise estimation of the relationship between ERCC1 and the prognosis and predictive response to chemotherapy of non-small cell lung cancer (NSCLC), a meta-analysis was performed. An electronic search of the PubMed and Embase database was performed. Hazard ratio (HR) for overall survival (OS) was pooled in early stage patients received surgery alone to analyze the prognosis of ERCC1 on NSCLC. HRs for OS in patients received surgery plus adjuvant chemotherapy and in patients received palliative chemotherapy and relative risk (RR) for overall response to chemotherapy were aggregated to analyze the prediction of ERCC1 on NSCLC. The pooled HR indicated that high ERCC1 levels were associated with longer survival in early stage patients received surgery alone (HR, 0.69; 95% confidence interval (CI), 0.58-0.83; P = 0.000). There was no difference in survival between high and low ERCC1 levels in patients received surgery plus adjuvant chemotherapy (HR, 1.41; 95% CI, 0.93-2.12; P = 0.106). However, high ERCC1 levels were associated with shorter survival and lower response to chemotherapy in advanced NSCLC patients received palliative chemotherapy (HR, 1.75; 95% CI, 1.39-2.22; P = 0.000; RR, 0.77; 95% CI, 0.64-0.93; P = 0.007; respectively). The meta-analysis indicated that high ERCC1 expression might be a favourable prognostic and a drug resistance predictive factor for NSCLC.
Collapse
|
44
|
Shao J, Zhang F, Bai Z, Wang C, Yuan Y, Wang W. Synthesis and antitumor activity of emodin quaternary ammonium salt derivatives. Eur J Med Chem 2012; 56:308-19. [PMID: 22901410 DOI: 10.1016/j.ejmech.2012.07.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 07/20/2012] [Accepted: 07/26/2012] [Indexed: 10/28/2022]
Abstract
A series of new emodin derivatives modified at the C-3 and the C-6 positions were synthesized, and evaluated for their anticancer activities in vitro and in vivo. Among them, Compounds 5g and 5h had more significant antiproliferative ability against HepG2, BGC-823, AGS cancer cell lines and low cytotoxicity to HELF normal cell line, respectively. Compounds 5g and 5h induced AGS cell cycle arrest at G0/G1 phase and induce apoptosis via activation of caspase-3 and caspase-9 enzyme. In vivo studies using H22 xenografts in Kunming mice were conducted with 5g and 5h. The results revealed that the medium dosage group (10 mg/kg) of 5g and the high dosage group (25 mg/kg) of 5h showed significant antitumor activity compared to the control group.
Collapse
Affiliation(s)
- Jingwei Shao
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Fuzhou University, Fujian 350108, PR China
| | | | | | | | | | | |
Collapse
|
45
|
Prodigiosin-induced cytotoxicity involves RAD51 down-regulation through the JNK and p38 MAPK pathways in human breast carcinoma cell lines. Toxicol Lett 2012; 212:83-9. [DOI: 10.1016/j.toxlet.2012.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/06/2012] [Accepted: 05/02/2012] [Indexed: 11/21/2022]
|
46
|
Kawashima A, Takayama H, Kawamura N, Doi N, Sato M, Hatano K, Nagahara A, Uemura M, Nakai Y, Nishimura K, Miyoshi S, Kawano K, Nishimura K, Nonomura N, Tsujimura A. Co-expression of ERCC1 and Snail is a prognostic but not predictive factor of cisplatin-based neoadjuvant chemotherapy for bladder cancer. Oncol Lett 2012; 4:15-21. [PMID: 22807952 DOI: 10.3892/ol.2012.689] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/02/2012] [Indexed: 11/05/2022] Open
Abstract
Neoadjuvant chemotherapy (NC) for bladder cancer has been reported to significantly improve the 5-year survival rate. The aim of the present study was to examine the roles of ERCC1 and Snail in determining the response to chemotherapy in bladder cancer treated with NC and radical cystectomy (RC). The expression of the Snail and ERCC1 proteins was determined by immunohistochemical staining of specimens obtained from 58 patients with bladder tumors treated with NC and RC. The correlation between clinical response and the expression of Snail and ERCC1 was investigated. Snail and ERCC1 were co-expressed in 24 (41.4%) of the 58 patients. A marked correlation was found between the expression of Snail and ERCC1 (P=0.001). The co-expression of Snail and ERCC1 was not able to predict pathological complete response (P=0.202). Results of the univariate analysis revealed that the co-expression of Snail and ERCC1 predicted shorter disease-free survival (DFS) and overall survival (OS) than the negative expression of Snail and/or ERCC1. Moreover, the co-expression of ERCC1 and Snail was the only predictive factor for both DFS (P=0.029) and OS (P=0.040). The expression of Snail was correlated with that of ERCC1 and the co-expression of Snail and ERCC1 was the only significant predictive factor of shorter DFS and OS in patients with bladder cancer treated with NC and RC.
Collapse
Affiliation(s)
- Atsunari Kawashima
- Department of Urology, Osaka University Graduate School of Medicine, Osaka
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Emodin loaded solid lipid nanoparticles: preparation, characterization and antitumor activity studies. Int J Pharm 2012; 430:238-46. [PMID: 22465546 DOI: 10.1016/j.ijpharm.2012.03.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 02/24/2012] [Accepted: 03/16/2012] [Indexed: 01/02/2023]
Abstract
The objective of the present study was to prepare and characterize emodin (EMO)-loaded solid lipid nanoparticles (E-SLNs) and evaluate their antitumor activity in vitro. EMO and pharmaceutical lipid material were used to prepare E-SLNs by high pressure homogenization (HPH). Poloxamer 188 and Tween 80 were used as surfactants. The physicochemical properties of the E-SLNs were investigated by particle size analysis, zeta potential measurement, drug entrapment efficiency (EE), stability and in vitro drug release behavior. The E-SLNs showed stable particle size at 28.6 ± 3.1 nm, ideal drug EE and relative long-term physical stability after being stored for 4 months. The drug release of E-SLNs could last 72 h and exhibited a sustained profile, which made it a promising vehicle for oral drug delivery. MTT assay showed that E-SLNs could significantly enhance the in vitro cytotoxicity against human breast cancer cell line MCF-7 and MDA-MB-231 cells compared to the EMO solution, while free EMO, blank SLNs (B-SLNs) and E-SLNs all showed no significant toxicity to human mammary epithelial line MCF-10A cells. Flow cytometric analysis demonstrated that E-SLNs also showed more significant cell cycle arrest effect in MCF-7 cells compared to bulk EMO solution. Hoechst 33342 staining and Annexin V-FITC/PI double staining further confirmed that E-SLNs induced higher apoptotic rates in MCF-7 cells, indicating that cell cycle arrest and apoptosis maybe the underlying mechanism of the enhanced cytotoxicity. Taken together, it seems that HPH was a simple, available and effective method for preparing high quality E-SLNs to enhance its aqueous solubility. Moreover, these results suggest that the delivery of EMO as lipid nanoparticles maybe a promising approach for cancer therapy.
Collapse
|
48
|
Ok S, Kim SM, Kim C, Nam D, Shim BS, Kim SH, Ahn KS, Choi SH, Ahn KS. Emodin inhibits invasion and migration of prostate and lung cancer cells by downregulating the expression of chemokine receptor CXCR4. Immunopharmacol Immunotoxicol 2012; 34:768-78. [PMID: 22299827 DOI: 10.3109/08923973.2012.654494] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Emodin (ED), an anthraquinone derivative, has been found to inhibit proliferation, induce apoptosis, suppress angiogenesis, impede metastasis, and enhance chemotherapy. However, the detailed mechanism of ED related to the regulation of CXC chemokine receptor-4 (CXCR4) gene expression that affects cellular migration and invasion in prostate and lung cancer cells are not fully understood. Recent evidence indicates that the CXCR4/CXCL12 axis is involved in promoting invasion and metastasis in tumors. Thus, novel agents that can downregulate CXCR4 expression have therapeutic potential in repressing cancer metastasis. Among ED and its derivatives, it is found that ED downregulated the expression of both CXCR4 and HER2 without affecting cell viability in tumor cells. The suppression of CXCR4 expression by ED was found to correlate with the inhibition of CXCL12-induced migration and invasion of both DU145 and A549 cells. Besides, neither proteasome inhibition nor lysosomal stabilization had any effect on ED-induced decrease in CXCR4 expression. The basic molecular mechanisms unveiled that the downregulation of CXCR4 was at the transcriptional level, as indicated by downregulation of mRNA expression and suppression of NF-κB activation. Overall, our findings suggest that ED is a novel blocker of CXCR4 expression and, thus, has enormous potential as a powerful therapeutic agent for metastatic cancer.
Collapse
Affiliation(s)
- Sooho Ok
- College of Oriental Medicine and Institute of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yang Z, Waldman AS, Wyatt MD. Expression and regulation of RAD51 mediate cellular responses to chemotherapeutics. Biochem Pharmacol 2011; 83:741-6. [PMID: 22222428 DOI: 10.1016/j.bcp.2011.12.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 12/14/2011] [Accepted: 12/16/2011] [Indexed: 02/07/2023]
Abstract
There is evidence that RAD51 expression associates with resistance to commonly used chemotherapeutics. Our previous work demonstrated that inhibitors of thymidylate synthase (TS) induced RAD51-dependent homologous recombination (HR), and depleting the RAD51 recombinase sensitized cells to TS inhibitors. In this study, the consequences of RAD51 over-expression were studied. Over-expression of wild-type RAD51 (∼6-fold above endogenous RAD51) conferred resistance to TS inhibitors. In contrast, over-expression of a mutant RAD51 (T309A) that is incapable of being phosphorylated rendered cells more chemosensitive. Moreover, over-expression of the T309A mutant acted in a dominant negative manner over endogenous RAD51 by causing the reduced localization of RAD51 foci following treatment with TS inhibitors. To measure the effect of mutant RAD51 on the cellular response to other DNA damaging chemotherapeutics, the topoisomerase poison etoposide was utilized. Cells over-expressing wild-type RAD51 showed reduced DNA strand breaks, while cells over-expressing the mutant RAD51 showed more than twice as many strand breaks, suggesting that the mutant RAD51 was actively inhibiting strand break resolution. To directly demonstrate an effect on HR, wild-type RAD51 and T309A mutant RAD51 were transiently expressed in HeLa cells that contained an HR reporter construct. HR events provoked by DNA breaks induced by the I-SceI endonuclease increased in cells expressing wild-type RAD51 and decreased in cells expressing the T309A mutant. Collectively, the data suggest that interference with the activation of RAD51-mediated HR represents a potentially useful anticancer target for combination therapies.
Collapse
Affiliation(s)
- Zhengguan Yang
- Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, United States
| | | | | |
Collapse
|
50
|
A Review of ERCC1 Gene in Bladder Cancer: Implications for Carcinogenesis and Resistance to Chemoradiotherapy. Adv Urol 2011; 2012:812398. [PMID: 22110495 PMCID: PMC3205704 DOI: 10.1155/2012/812398] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 08/30/2011] [Indexed: 02/06/2023] Open
Abstract
The excision repair cross-complementing group 1 (ERCC1) gene performs a critical incision step in DNA repair and is reported to be correlated with carcinogenesis and resistance to drug or ionizing radiation therapy. We reviewed the correlation between ERCC1 and bladder cancer. In carcinogenesis, several reports discussed the relation between ERCC1 single nucleotide polymorphisms and carcinogenesis in bladder cancer only in case-control studies. Regarding the relation between ERCC1 and resistance to chemoradiotherapy, in vitro and clinical studies indicate that ERCC1 might be related to resistance to radiation therapy rather than cisplatin therapy. It is controversial whether ERCC1 predicts prognosis of bladder cancer treated with cisplatin-based chemotherapy. Tyrosine kinase receptors or endothelial-mesenchymal transition are reported to regulate the expression of ERCC1, and further study is needed to clarify the mechanism of ERCC1 expression and resistance to chemoradiotherapy in vitro and to discover novel therapies for advanced and metastatic bladder cancer.
Collapse
|