1
|
Guo Z, Meng Y, Li X, Li J, Zhou S, Feng R, Wu W, Xu M, Liu J, Zeng X, Zhao W, Zhong H. Single molecule recognition of CD95 receptors on the surface of HepG2 cells under the curcumin. J Pharm Biomed Anal 2025; 263:116917. [PMID: 40300310 DOI: 10.1016/j.jpba.2025.116917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Hepatocellular carcinoma (HCC) is a serious concern worldwide. The published reports showed that aberrant CD95 receptor expression plays a critical role in apoptosis in liver cancer. While curcumin has shown promise in inducing apoptosis in liver cancer cells, its direct effects on CD95 expression during this process have not been thoroughly investigated. This study aims to quantitatively assess the expression of the CD95 receptor in HepG2 cells treated with different concentrations of curcumin using techniques such as fluorescence staining, single-molecule force spectroscopy (SMFS), and single-molecule recognition imaging (SMRI). Fluorescence staining results indicate a significant increase in CD95 expression following curcumin treatment. For the first time, SMFS and SMRI techniques were used to directly reveal the binding sites of CD95 on the cell membrane, with the number of binding sites increasing as the curcumin concentration increased. Additionally, the binding force between an antibody-modified probe and CD95 was strengthened with curcumin treatment, suggesting that curcumin enhances both the quantity and affinity of CD95 binding sites. This study provides new insights into curcumin-induced CD95-mediated apoptosis in liver cancer cells and highlights the potential of AFM techniques for investigating drug mechanisms. Overall, these findings may inform innovative therapeutic strategies for liver cancer and improve drug design processes.
Collapse
Affiliation(s)
- Zeling Guo
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, PR China
| | - Yu Meng
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China
| | - Xinyu Li
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China
| | - Jiangting Li
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China
| | - Shang Zhou
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China
| | - Rongrong Feng
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China
| | - Weiting Wu
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China
| | - Mingjing Xu
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China
| | - Jinhao Liu
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China
| | - Xiangfu Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China.
| | - Weidong Zhao
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China.
| | - Haijian Zhong
- Key Laboratory for Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, PR China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, PR China.
| |
Collapse
|
2
|
Wen Y, Song N, Peng Y, Wu W, Lin Q, Cui M, Li R, Yu Q, Wu S, Liang Y, Tian W, Meng Y. Radiofrequency enhances drug release from responsive nanoflowers for hepatocellular carcinoma therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:569-579. [PMID: 38887527 PMCID: PMC11181167 DOI: 10.3762/bjnano.15.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 06/20/2024]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor and the third leading cause of cancer death worldwide. Most patients are diagnosed at an advanced stage, and systemic chemotherapy is the preferred treatment modality for advanced HCC. Curcumin (CUR) is a polyphenolic antineoplastic drug with low toxicity obtained from plants. However, its low bioavailability and poor solubility limit its functionality. In this study, radiofrequency- (RF) enhanced responsive nanoflowers (NFs), containing superparamagnetic ferric oxide nanoclusters (Fe3O4 NCs), - CUR layer, - and MnO2 (CUR-Fe@MnO2 NFs), were verified to have a thermal therapeutic effect. Transmission electron microscopy was used to characterize the CUR-Fe@MnO2 NFs, which appeared flower-like with a size of 96.27 nm. The in vitro experimental data showed that RF enhanced the degradation of CUR-Fe@MnO2 NFs to release Mn2+ and CUR. The cytotoxicity test results indicated that after RF heating, the CUR-Fe@MnO2 NFs significantly suppressed HCC cell proliferation. Moreover, CUR-Fe@MnO2 NFs were effective T 1/T 2 contrast agents for molecular magnetic resonance imaging due to the release of Mn2+ and Fe3O4 NCs.
Collapse
Affiliation(s)
- Yanyan Wen
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ningning Song
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yueyou Peng
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Weiwei Wu
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Qixiong Lin
- The Ninth Clinical Medical School of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Minjie Cui
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Rongrong Li
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Qiufeng Yu
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
- Department of Medical Imaging, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Sixue Wu
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yongkang Liang
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
- Department of Medical Imaging, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Wei Tian
- Department of General Surgery, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, China
| | - Yanfeng Meng
- Department of Radiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| |
Collapse
|
3
|
Hsueh KC, Ju PC, Hsieh YH, Su SC, Yeh CB, Lin CW. HO-3867, a curcumin analog, elicits cell apoptosis and p38-mediated caspase activation in hepatocellular carcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:794-802. [PMID: 37782689 DOI: 10.1002/tox.23977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
HO-3867, a synthetic curcumin analog, has displayed various tumor-suppressive characteristics and improved bioabsorption over its parent compound. However, its influences on the development of hepatocellular carcinoma (HCC) are poorly defined. To address this, we tested the anticarcinogenic impact of HO-3867 and investigated the underlying mechanisms in fighting liver cancer. Our result demonstrated that HO-3867 reduced the viability of HCC cells, accompanied by promotion of cell cycle arrest at the sub-G1 stage and apoptotic responses. Furthermore, a distinctive profile of apoptosis associated proteins, encompassing elevated heme oxygenase-1 (HO-1) level and caspase activation, was detected in HO-3867-stimulated HCC cells. In addition, such HO-3867-mediated elevation in caspase activation was dampened by pharmacological suppression of p38 activities. Taken together, our findings unveiled that HO-3867 triggered cell cycle arrest and apoptotic events in liver cancer, involving a p38-mediated activation of caspase cascades. These data highlighted a usefulness of curcumin or its analogs on the management of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Kuan-Chun Hsueh
- Division of General Surgery, Department of Surgery, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Po-Chung Ju
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chao-Bin Yeh
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Suwannasom N, Sriaksorn N, Thepmalee C, Khoothiam K, Prapan A, Bäumler H, Thephinlap C. Curcumin-loaded albumin submicron particles with potential as a cancer therapy: an in vitro study. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:1127-1140. [PMID: 38034473 PMCID: PMC10682534 DOI: 10.3762/bjnano.14.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023]
Abstract
Curcumin (CUR), a polyphenolic compound, shows promising biological properties, particularly antioxidant activity. However, its medical applications are limited due to its low water solubility, bioavailability, and pH-instability. CUR-loaded albumin microparticles (CUR-HSA-MPs) of submicron size in the range of 800 to 900 nm and a zeta potential of -15 mV were prepared. The CUR loading efficiency was up to 65%. A maximum release of 37% of the encapsulated CUR was observed within 6 h when the CUR-HSA-MPs were dispersed in 50% ethanol in PBS at pH 7, while in RPMI 1640 medium the release was 7%. This demonstrates a sustainable release. The in vitro cytotoxicity of CUR-HSA-MPs showed promising anticancer potential against human hepatocellular carcinoma (Huh-7) and human breast adenocarcinoma (MCF-7) cell lines, although this effect was less pronounced in human dermal fibroblasts (HDFB) and human cholangiocyte (MMN) cell lines. Confocal microscopy was used to confirm the uptake of CUR-HSA-MPs by cancer cells. Our studies revealed that HSA-MPs are potentially promising vehicles for increasing the solubility and bioavailability of CUR.
Collapse
Affiliation(s)
- Nittiya Suwannasom
- Division of Biochemistry, School of Medical Sciences, University of Phayao 56000, Thailand
| | - Netsai Sriaksorn
- Division of Biochemistry, School of Medical Sciences, University of Phayao 56000, Thailand
| | - Chutamas Thepmalee
- Division of Biochemistry, School of Medical Sciences, University of Phayao 56000, Thailand
| | - Krissana Khoothiam
- Department of Radiological Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Ausanai Prapan
- Division of Microbiology, School of Medical Sciences, University of Phayao 56000, Thailand
| | - Hans Bäumler
- Institute of Transfusion Medicine, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Chonthida Thephinlap
- Division of Biochemistry, School of Medical Sciences, University of Phayao 56000, Thailand
| |
Collapse
|
5
|
Shamsnia HS, Roustaei M, Ahmadvand D, Butler AE, Amirlou D, Soltani S, Momtaz S, Jamialahmadi T, Abdolghaffari AH, Sahebkar A. Impact of curcumin on p38 MAPK: therapeutic implications. Inflammopharmacology 2023; 31:2201-2212. [PMID: 37498375 DOI: 10.1007/s10787-023-01265-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 07/28/2023]
Abstract
Curcumin (diferuloylmethane) is a herbal remedy which possesses numerous biological attributes including anti-inflammatory, anti-oxidant and anti-cancer properties. Curcumin has been shown to impact a number of signaling pathways including nuclear factor kappa B (NF-KB), reactive oxygen species (ROS), Wingless/Integrated (Wnt), Janus kinase-signal transducer and activator of mitogen-activated protein kinase (MAPK) and transcription (JAK/STAT). P38 belongs to the MAPKs, is known as a stress-activated MAPK and is involved in diverse biological responses. P38 is activated in various signaling cascades. P38 plays a role in inflammation, cell differentiation, proliferation, motility and survival. This cascade can serve as a therapeutic target in many disorders. Extensive evidence confirms that curcumin impacts the P38 MAPK signaling pathway, through which it exerts anti-inflammatory, neuroprotective, and apoptotic effects. Hence, curcumin can positively affect inflammatory disorders and cancers, as well as to increase glucose uptake in cells. This review discusses the pharmacological and therapeutic effects of curcumin as effected through p38 MAPK.
Collapse
Affiliation(s)
- Hedieh Sadat Shamsnia
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahtab Roustaei
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Danial Ahmadvand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Dorsa Amirlou
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sanam Soltani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Medicine, The University of Western Australia, Perth, Australia.
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Thepmalee C, Sawasdee N, Jenkham P, Thephinlap C, Khoothiam K, Suwannasom N, Chokchaisiri R, Panya A, Yenchitsomanus PT. Anti-cancer effect of a phytochemical compound - 7R-acetylmelodorinol - against triple-negative breast cancer cells. Biomed Pharmacother 2023; 166:115286. [PMID: 37573655 DOI: 10.1016/j.biopha.2023.115286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype currently lacking effective treatment options. Consequently, novel and effective drugs or compounds are urgently needed to treat TNBC. Therefore, this study aimed to evaluate the potential of 7R-acetylmelodorinol (7R-AMDL), a phytochemical compound isolated from Xylopia pierrei Hance, a plant found in Thailand, as a novel therapeutic agent for TNBC. MTT and clonogenic assays showed that 7R-AMDL significantly reduced the survival of breast cancer cell lines, with a markedly potent effect on MDA-MB-231 cells. Flow cytometry showed that treating MDA-MB-231 cells with 7R-AMDL at the concentration of dose 8 µM significantly increased early and late apoptosis after 24 and 48 h compared to the control group (p < 0.0001). The highest tested 7R-AMDL dose upregulated the death receptors and their ligands, with extrinsic and intrinsic apoptosis pathways significantly activated via the caspase cascade, compared to the untreated group (p < 0.05). In addition, immunoblots showed decreased BCL2-like 1 (BCL2L1/Bcl-xL) expression (p < 0.0001). Furthermore, wound healing and Transwell assays showed that at a non-cytotoxic dose (≤4 µM), 7R-AMDL significantly inhibited the MDA-MB-231 cell migration and invasion. This reduction in cell migration was associated with decreased matrix metallopeptidase 9 (MMP-9) expression (p < 0.01) and nuclear factor kappa B (NF-κB) activation (p < 0.05). Altogether, 7R-AMDL has anti-cancer effects against TNBC and the potential to be further developed and evaluated for treating this disease.
Collapse
Affiliation(s)
- Chutamas Thepmalee
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao 56000, Thailand; Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand.
| | - Nunghathai Sawasdee
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Phanitaporn Jenkham
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao 56000, Thailand; Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Chonthida Thephinlap
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao 56000, Thailand; Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Krissana Khoothiam
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao 56000, Thailand; Division of Microbiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Nittiya Suwannasom
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao 56000, Thailand; Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | | | - Aussara Panya
- Cell Engineering for Cancer Therapy Research Group, Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
7
|
Chimento A, D’Amico M, De Luca A, Conforti FL, Pezzi V, De Amicis F. Resveratrol, Epigallocatechin Gallate and Curcumin for Cancer Therapy: Challenges from Their Pro-Apoptotic Properties. Life (Basel) 2023; 13:life13020261. [PMID: 36836619 PMCID: PMC9962739 DOI: 10.3390/life13020261] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Plant-derived bioactive compounds are gaining wide attention for their multiple health-promoting activities and in particular for their anti-cancer properties. Several studies have highlighted how they can prevent cancer initiation and progression, improve the effectiveness of chemotherapy, and, in some cases, limit some of the side effects of chemotherapy agents. In this paper, we provide an update of the literature on the anti-cancer effects of three extensively studied plant-derived compounds, namely resveratrol, epigallocatechin gallate, and curcumin, with a special focus on the anti-cancer molecular mechanisms inducing apoptosis in the major types of cancers globally.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria D’Amico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Arianna De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
- Correspondence: ; Tel.: +39-0984-496204
| |
Collapse
|
8
|
The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy. Int J Mol Sci 2023; 24:ijms24021680. [PMID: 36675194 PMCID: PMC9863215 DOI: 10.3390/ijms24021680] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Various literature data show how a diet rich in vegetables could reduce the incidence of several cancers due to the contribution of the natural polyphenols contained in them. Polyphenols are attributed multiple pharmacological actions such as anti-inflammatory, anti-oxidant, antibiotic, antiseptic, anti-allergic, cardioprotective and even anti-tumor properties. The multiple mechanisms involved in their anti-tumor action include signaling pathways modulation associated with cell proliferation, differentiation, migration, angiogenesis, metastasis and cell death. Since the dysregulation of death processes is involved in cancer etiopathology, the natural compounds able to kill cancer cells could be used as new anticancer agents. Apoptosis, a programmed form of cell death, is the most potent defense against cancer and the main mechanism used by both chemotherapy agents and polyphenols. The aim of this review is to provide an update of literature data on the apoptotic molecular mechanisms induced by some representative polyphenol family members in cancer cells. This aspect is particularly important because it may be useful in the design of new therapeutic strategies against cancer involving the polyphenols as adjuvants.
Collapse
|
9
|
Sudhesh Dev S, Zainal Abidin SA, Farghadani R, Othman I, Naidu R. Receptor Tyrosine Kinases and Their Signaling Pathways as Therapeutic Targets of Curcumin in Cancer. Front Pharmacol 2021; 12:772510. [PMID: 34867402 PMCID: PMC8634471 DOI: 10.3389/fphar.2021.772510] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are transmembrane cell-surface proteins that act as signal transducers. They regulate essential cellular processes like proliferation, apoptosis, differentiation and metabolism. RTK alteration occurs in a broad spectrum of cancers, emphasising its crucial role in cancer progression and as a suitable therapeutic target. The use of small molecule RTK inhibitors however, has been crippled by the emergence of resistance, highlighting the need for a pleiotropic anti-cancer agent that can replace or be used in combination with existing pharmacological agents to enhance treatment efficacy. Curcumin is an attractive therapeutic agent mainly due to its potent anti-cancer effects, extensive range of targets and minimal toxicity. Out of the numerous documented targets of curcumin, RTKs appear to be one of the main nodes of curcumin-mediated inhibition. Many studies have found that curcumin influences RTK activation and their downstream signaling pathways resulting in increased apoptosis, decreased proliferation and decreased migration in cancer both in vitro and in vivo. This review focused on how curcumin exhibits anti-cancer effects through inhibition of RTKs and downstream signaling pathways like the MAPK, PI3K/Akt, JAK/STAT, and NF-κB pathways. Combination studies of curcumin and RTK inhibitors were also analysed with emphasis on their common molecular targets.
Collapse
Affiliation(s)
- Sareshma Sudhesh Dev
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Malaysia
| | - Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Malaysia
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Malaysia
| |
Collapse
|
10
|
Nguyen HT, Kacimi SEO, Nguyen TL, Suman KH, Lemus-Martin R, Saleem H, Do DN. MiR-21 in the Cancers of the Digestive System and Its Potential Role as a Diagnostic, Predictive, and Therapeutic Biomarker. BIOLOGY 2021; 10:biology10050417. [PMID: 34066762 PMCID: PMC8151274 DOI: 10.3390/biology10050417] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs. They can regulate the expression of their target genes, and thus, their dysregulation significantly contributes to the development of cancer. Growing evidence suggests that miRNAs could be used as cancer biomarkers. As an oncogenic miRNA, the roles of miR-21 as a diagnostic and prognostic biomarker, and its therapeutic applications have been extensively studied. In this review, the roles of miR-21 are first demonstrated via its different molecular networks. Then, a comprehensive review on the potential targets and the current applications as a diagnostic and prognostic cancer biomarker and the therapeutic roles of miR-21 in six different cancers in the digestive system is provided. Lastly, a brief discussion on the challenges for the use of miR-21 as a therapeutic tool for these cancers is added.
Collapse
Affiliation(s)
- Ha Thi Nguyen
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam;
- Faculty of Medicine, Duy Tan University, Danang 550000, Vietnam
| | | | - Truc Ly Nguyen
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea;
| | - Kamrul Hassan Suman
- Department of Fisheries Biology & Aquatic Environment, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | | | - Humaira Saleem
- Jamil–ur–Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan;
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS B2N5E3, Canada
- Correspondence: ; Tel.: +1-819-571-5310
| |
Collapse
|
11
|
Son HK, Kim D, Lim Y, Kim J, Park I. A novel TGF-β receptor II mutation (I227T/N236D) promotes aggressive phenotype of oral squamous cell carcinoma via enhanced EGFR signaling. BMC Cancer 2020; 20:1163. [PMID: 33246423 PMCID: PMC7694911 DOI: 10.1186/s12885-020-07669-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 11/20/2020] [Indexed: 01/02/2023] Open
Abstract
Background Transforming growth factor-β (TGF-β) signaling is a double-edged sword in cancer development and progression. TGF-β signaling plays a tumor suppressive role during the early stages of tumor development but promotes tumor progression in later stages. We have previously identified various mutations of TGF-β receptor II (TβRII) in human oral squamous cell carcinoma (OSCC) samples. In the present study we analyzed I227T/N236D mutation of TβRII, which was detected in the metastatic lymph node of an OSCC patient. Methods The effect of I227T/N236D TβRII mutation on transcriptional activities was measured using DR26 cells, which lack functional TβRII. HSC2 human OSCC cells stably expressing wild-type and I227T/N236D mutant TβRII were generated and used to examine the effect of I227T/N236D TβRII mutation on xenograft tumor growth, in vitro cell proliferation, apoptosis, migration, and invasion. Results The I227T/N236D mutation of TβRII upregulated TGF-β signaling and promoted xenograft tumor growth when compared with the wild-type, without affecting the in vitro proliferative capacities. To delineate the differences in proliferative capacities in vivo and in vitro, the apoptotic and survival signals were analyzed following curcumin treatment. Concomitant with apoptotic induction, epidermal growth factor receptor (EGFR) activation was observed upon curcumin treatment, which was further activated in I227T/N236D mutant transfectant cells when compared with wild-type cells. Enhanced EGFR activation correlated with cell survival and apoptotic resistance. Enhanced migratory and invasive capabilities of I227T/N236D mutant cells also depended on EGFR signaling. Conclusions These results suggest that enhanced EGFR signaling via upregulated TGF-β signaling shifted the balance toward survival and promoted cell migration and invasion in I227T/N236D mutant cells, elucidating the role of I227T/N236D mutation of TβRII in OSCC progression.
Collapse
Affiliation(s)
- Hwa-Kyung Son
- Department of Dental Hygiene, Yeungnam University College, Daegu, 42415, Republic of Korea
| | - Dokyeong Kim
- Department of Dental Hygiene, Jeonju Kijeon College, Jeonju, 54989, Republic of Korea.,Department of Oral Pathology, Oral Cancer Research Institute, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Yongwoon Lim
- Department of Biochemistry, Department of Biomedical Sciences, Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Jin Kim
- Department of Oral Pathology, Oral Cancer Research Institute, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Iha Park
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
12
|
Qu J, Lu W, Chen M, Gao W, Zhang C, Guo B, Yang J. Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer. Exp Ther Med 2020; 20:18. [PMID: 32934683 PMCID: PMC7471865 DOI: 10.3892/etm.2020.9145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 01/17/2020] [Indexed: 01/27/2023] Open
Abstract
The development of an effective therapeutic intervention for liver cancer is a worldwide challenge that remains to be adequately addressed. Of note, TP53, which encodes the p53 protein, is an important tumor suppressor gene, 61% of TP53 is functionally inactivated in liver cancer. Recombinant human adenovirus p53 (rAd-p53) is the first commercial product that has been used for gene therapy. In the present study, the combined mechanistic effects of rAd-p53 and curcumin, a naturally occurring compound with previously reported anti-inflammatory, antioxidant and anti-cancer properties, were assessed in liver cancer cells, using HepG2 cells as the model cell line. The administration of either curcumin or rAd-p53 promoted apoptosis, suppressed epithelial-mesenchymal transition (EMT) and blocked G2/M phase progression in HepG2 cells, which were potentiated further when both agents were applied together. Combined rAd-p53 and curcumin treatment resulted in higher p53 (P<0.01) and p21 (P<0.01) expression compared with rAd-p53 or curcumin were added alone, suggesting an additive effect on TP53 expression. Additionally, curcumin and rAd-p53 were demonstrated to regulate the activation of mitogen-activated protein kinases (MAPKs) ERK1/2, p38 MAPK and JNK. These results indicated that the combination of rAd-p53 with curcumin synergistically potentiates apoptosis and inhibit EMT compared with either rAd-p53 or curcumin treatment alone via the regulation of TP53 regulation. Mechanistically, this effect on TP53 expression may involve the ERK1/2, p38 MAPK and JNK signaling pathways. The current study provides new insights that can potentially advance the development of therapeutic strategies for liver cancer treatment.
Collapse
Affiliation(s)
- Juan Qu
- Department of Gastroenterology, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Wei Lu
- Department of Gastroenterology, Tianjin Cancer Hospital, Tianjin 300060, P.R. China
| | - Ming Chen
- Department of Hepatopathy and Hepatic Oncology, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Wei Gao
- Department of Hepatopathy and Hepatic Oncology, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Cong Zhang
- Department of Hepatopathy and Hepatic Oncology, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Bin Guo
- College of Acu-moxibustion and Massage, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jizhi Yang
- Department of Traditional Chinese Medicine, Chentangzhuang Street Health Service Center, Tianjin 300222, P.R. China
| |
Collapse
|
13
|
Curcumin-loaded nanostructured lipid carrier induced apoptosis in human HepG2 cells through activation of the DR5/caspase-mediated extrinsic apoptosis pathway. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2020; 70:227-237. [PMID: 31955141 DOI: 10.2478/acph-2020-0003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/19/2019] [Indexed: 01/19/2023]
Abstract
Curcumin is a lipophilic anti-cancer compound extracted from turmeric. Our previous study demonstrated that the curcumin-loaded nanostructured lipid carrier (Cur-NLC) exhibits superior anti-cancer activity in inhibiting proliferation as well as inducing apoptosis of human HepG2 cells compared to native curcumin. This study aims to unveil the mechanisms underlying the pro-apoptotic effect of Cur-NLC on HepG2 cells. Evidence indicates that low expression of death receptors (DRs) on cancer cell membranes leads to attenuated apoptosis signaling. This study showed that Cur-NLC significantly increased total expression of DR5 protein while simultaneously upregulated cell membrane expression of DR5. Cur-NLC significantly increased caspase-8 and caspase-3 activities, accompanied by increased apoptosis. Furthermore, enhanced apoptosis was inhibited in the presence of a pan-caspase inhibitor, Z-VAD-FMK. Therefore, Cur-NLC induced activation of the extrinsic apoptosis pathway via modulating the DR5/caspase-8/-3 mediated apoptosis pathway in HepG2 cells, suggesting that Cur-NLC is a promising therapeutic agent or supplement for the treatment of hepatocellular carcinoma.
Collapse
|
14
|
Gao J, Dai C, Yu X, Yin XB, Zhou F. Long noncoding RNA LINC00324 exerts protumorigenic effects on liver cancer stem cells by upregulating fas ligand via PU box binding protein. FASEB J 2020; 34:5800-5817. [PMID: 32128906 DOI: 10.1096/fj.201902705rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) represents a major cause of cancer death, but the molecular mechanism for its development has not yet been well characterized. Long noncoding RNAs (lncRNAs) are involved in a wide range of biological processes via their roles as oncogenes or tumor suppressor genes. The present study aimed to elucidate the role of LINC00324 in HCC through its interaction with Fas ligand (FasL). Initially, microarray-based gene expression profiling of HCC was employed to identify differentially expressed genes. Next, the expression of LINC00324 in HCC tissues and liver cancer stem cell (LCSC) lines was examined using RT-qPCR. Then, the interaction among LINC00324, PU box binding protein (PU.1) and FasL was identified with RIP, ChIP and dual-luciferase reporter gene assays. The effect of LINC00324 on viability, proliferation, migration, invasion, and apoptosis as well as the tumorigenesis of transfected cells was examined with gain- and loss-of-function experiments. LINC00324 and FasL were highly expressed in HCC. LINC00324 regulated FasL expression via interaction with PU.1. Silencing of LINC00324 or FasL suppressed expression of stemness-related genes, cell viability, proliferation, migration, invasion, self-renewal, and tumorigenesis, but enhanced cell apoptosis. Taken together, LINC00324 promotes the expression of FasL through the recruitment of PU.1, which ultimately maintains the biological properties of LCSCs, thus, highlighting LINC00324 as a promising therapeutic candidate for HCC.
Collapse
Affiliation(s)
- Jun Gao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Chao Dai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xin Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiang-Bao Yin
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Fan Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
15
|
Wu LY, Chen CW, Chen LK, Chou HY, Chang CL, Juan CC. Curcumin Attenuates Adipogenesis by Inducing Preadipocyte Apoptosis and Inhibiting Adipocyte Differentiation. Nutrients 2019; 11:nu11102307. [PMID: 31569380 PMCID: PMC6836120 DOI: 10.3390/nu11102307] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/09/2019] [Accepted: 09/10/2019] [Indexed: 12/23/2022] Open
Abstract
Patients with metabolic syndrome are at an increased risk of developing type 2 diabetes and cardiovascular diseases. The principal risk factor for development of metabolic syndrome is obesity, defined as a state of pathological hyperplasia or/and hypertrophy of adipose tissue. The number of mature adipocytes is determined by adipocyte differentiation from preadipocytes. The purpose of the present study is to investigate the effects of curcumin on adipogenesis and the underlying mechanism. To examine cell toxicity of curcumin, 3T3-L1 preadipocytes were treated with 0–50 µM curcumin for 24, 48, or 72 h, then cell viability was measured using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. The effect of curcumin on the cell cycle was determined by flow cytometry. Curcumin-induced cell apoptosis was determined by the TUNEL assay and curcumin-induced caspase activation was measured by immunoblotting. The effect of curcumin on adipocyte differentiation was determined by measuring mitotic clonal expansion (MCE), expression of adipogenic transcription factors, and lipid accumulation. Results showed the viability of preadipocytes was significantly decreased by treatment with 30 µM curcumin, a concentration that caused apoptosis in preadipocytes, as assessed by the TUNEL assay, and caused activation of caspases 8, 9, and 3. A non-cytotoxic dose of curcumin (15 µM) inhibited MCE, downregulated the expression of PPARγ and C/EBPα, prevented differentiation medium-induced β-catenin downregulation, and decreased the lipid accumulation in 3T3-L1 adipocytes. In conclusion, our data show that curcumin can induce preadipocyte apoptosis and inhibit adipocyte differentiation, leading to suppression of adipogenesis.
Collapse
Affiliation(s)
- Liang-Yi Wu
- Department of Bioscience Technology, College of Science, Chung-Yuan Christian University, Chung Li 32023, Taiwan.
| | - Chien-Wei Chen
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan.
| | - Luen-Kui Chen
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Hsiang-Yun Chou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Chih-Ling Chang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Chi-Chang Juan
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| |
Collapse
|
16
|
Du X, Hong L, Sun L, Sang H, Qian A, Li W, Zhuang H, Liang H, Song D, Li C, Wang W, Li X. miR-21 induces endothelial progenitor cells proliferation and angiogenesis via targeting FASLG and is a potential prognostic marker in deep venous thrombosis. J Transl Med 2019; 17:270. [PMID: 31416448 PMCID: PMC6694687 DOI: 10.1186/s12967-019-2015-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 08/04/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Deep venous thrombosis (DVT) of lower extremities is a common thrombotic disease, occurring either in isolation or as a complication of other diseases or procedures. MiR-21 is one of important microRNAs which play critical role in various cellular function. This study aim to determine the effect of miR-21 on endothelial progenitor cells (EPCs) and its role in predicting prognosis of DVT. METHODS EPCs was isolated from DVT models and control subjects. miR-21 expression was confirmed by RT-PCR. Potential target mRNA was predicted by bioinformatics analysis. EPCs biological functions were examined by CCK-8 and tube formation assay. Besides, miR-21 expression was determined in DVT patients to investigate the correlation between miR-21 expression and prognosis of DVT. Cox proportional hazard regression analyses were also performed to reveal the risk factors associated with prognosis. RESULTS Here, we found miR-21 was downregulated in EPCs of DVT model rats. Increased miR-21 expression promoted proliferation and angiogenesis of EPCs. Moreover, we demonstrated that FASLG was a target of miR-21 and revealed that FASLG knockdown inhibited function of EPCs. Upregulation of miR-21 led to thrombus resolution in a rat model of venous thrombosis. In addition, lower expression level of miR-21 in DVT patients was associated with an increase of recurrent DVT and post thrombotic syndrome (PTS). Furthermore, Cox proportional hazard regression analyses demonstrated miR-21 expression level as an independent predictor of recurrence of DVT. CONCLUSIONS Our data revealed a role of miR-21 in regulating biological function of EPCs and could be a predictor for recurrent DVT or PTS.
Collapse
Affiliation(s)
- Xiaolong Du
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Lei Hong
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.,Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Lili Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.,Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Hongfei Sang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Aiming Qian
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wendong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Hao Zhuang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Huoqi Liang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Dandan Song
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Chenglong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Xiaoqiang Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| |
Collapse
|
17
|
Yue SJ, Zhang PX, Zhu Y, Li NG, Chen YY, Li JJ, Zhang S, Jin RY, Yan H, Shi XQ, Tang YP, Duan JA. RETRACTED: A Ferulic Acid Derivative FXS-3 Inhibits Proliferation and Metastasis of Human Lung Cancer A549 Cells via Positive JNK Signaling Pathway and Negative ERK/p38, AKT/mTOR and MEK/ERK Signaling Pathways. Molecules 2019; 24:E2165. [PMID: 31181779 PMCID: PMC6600170 DOI: 10.3390/molecules24112165] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/28/2019] [Accepted: 06/01/2019] [Indexed: 11/17/2022] Open
Abstract
Lung cancer is one of the most common malignancies and is an increasing cause of cancer-related deaths. In our previous study, a series of ferulic acid (FA) derivatives were designed and synthesized; they exhibited positive anti-cancer activities, especially for a compound labelled FXS-3. In this study, a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed, wherein it revealed the inhibitory effect of FXS-3 on the proliferation and metastasis of human lung cancer A549 cells. The further flow cytometry assay showed that FXS-3 induced apoptosis of A549 cells induced cell cycle arrest at the G0/G1 phase. The trans-well migration and Matrigel invasion assays revealed that FXS-3 inhibited the migration and invasion of A549 cells. By the western blotting analysis, FXS-3 increased the expression of B-cell lymphoma-2 (Bcl-2) associated X protein (Bax)/Bcl-2 ratio, inhibited matrix metalloproteinase (MMP)-2 and MMP-9, and regulated the extracellular signal-regulated kinase (ERK)/p38, c-Jun N-terminal kinase (JNK), protein kinase B (AKT)/mechanistic target of rapamycin (mTOR), as well as mitogen-activated protein kinase (MEK)/ERK signaling pathways. The subsequent A549 xenograft-bearing mouse model and tail vein injection of A549 cells induced pulmonary tumor metastasis model showed that FXS-3 significantly restrained the tumor growth and metastasis. In conclusion, FXS-3 might inhibit proliferation and metastasis of human lung cancer A549 cells by positively regulating JNK signaling pathway and negativly regulating ERK/p38, AKT/mTOR, and MEK/ERK signaling pathways, which provides important scientific basis for the development of anti-cancer drugs about FA derivatives.
Collapse
Affiliation(s)
- Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Peng-Xuan Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Yue Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Nian-Guang Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Yan-Yan Chen
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Jia-Jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Sai Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Ru-Yi Jin
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Hao Yan
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Xu-Qin Shi
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| |
Collapse
|
18
|
Extrinsic or Intrinsic Apoptosis by Curcumin and Light: Still a Mystery. Int J Mol Sci 2019; 20:ijms20040905. [PMID: 30791477 PMCID: PMC6412849 DOI: 10.3390/ijms20040905] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 02/03/2023] Open
Abstract
Curcumin—a rhizomal phytochemical from the plant Curcuma longa—is well known to inhibit cell proliferation and to induce apoptosis in a broad range of cell lines. In previous studies we showed that combining low curcumin concentrations and subsequent ultraviolet A radiation (UVA) or VIS irradiation induced anti-proliferative and pro-apoptotic effects. There is still debate whether curcumin induces apoptosis via the extrinsic or the intrinsic pathway. To address this question, we investigated in three epithelial cell lines (HaCaT, A431, A549) whether the death receptors CD95, tumor necrosis factor (TNF)-receptor I and II are involved in apoptosis induced by light and curcumin. Cells were incubated with 0.25–0.5 µg/mL curcumin followed by irradiation with 1 J/cm2 UVA. This treatment was combined with inhibitors specific for distinct membrane-bound death receptors. After 24 h apoptosis induction was monitored by quantitative determination of cytoplasmic histone-associated-DNA-fragments. Validation of our test system showed that apoptosis induced by CH11 and TNF-α could be completely inhibited by their respective antagonists. Interestingly, apoptosis induced by curcumin/light treatment was reversed by none of the herein examined death receptor antagonists. These results indicate a mechanism of action independent from classical death receptors speaking for intrinsic activation of apoptosis. It could be speculated that a shift in cellular redox balance might prompt the pro-apoptotic processes.
Collapse
|
19
|
Mortezaee K, Salehi E, Mirtavoos-Mahyari H, Motevaseli E, Najafi M, Farhood B, Rosengren RJ, Sahebkar A. Mechanisms of apoptosis modulation by curcumin: Implications for cancer therapy. J Cell Physiol 2019; 234:12537-12550. [PMID: 30623450 DOI: 10.1002/jcp.28122] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Cancer incidences are growing and cause millions of deaths worldwide. Cancer therapy is one of the most important challenges in medicine. Improving therapeutic outcomes from cancer therapy is necessary for increasing patients' survival and quality of life. Adjuvant therapy using various types of antibodies or immunomodulatory agents has suggested modulating tumor response. Resistance to apoptosis is the main reason for radioresistance and chemoresistance of most of the cancers, and also one of the pivotal targets for improving cancer therapy is the modulation of apoptosis signaling pathways. Apoptosis can be induced by intrinsic or extrinsic pathways via stimulation of several targets, such as membrane receptors of tumor necrosis factor-α and transforming growth factor-β, and also mitochondria. Curcumin is a naturally derived agent that induces apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing reactive oxygen species (ROS) production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of NF-κB and COX-2, which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis PI3K signaling and increase the expression of MAPKs to induce endogenous production of ROS. In this paper, we aimed to review the molecular mechanisms of curcumin-induced apoptosis in cancer cells. This action of curcumin could be applicable for use as an adjuvant in combination with other modalities of cancer therapy including radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Zhang X, Dai F, Chen J, Xie X, Xu H, Bai C, Qiao W, Shen W. Antitumor effect of curcumin liposome after transcatheter arterial embolization in VX2 rabbits. Cancer Biol Ther 2019; 20:642-652. [PMID: 30621501 PMCID: PMC6606009 DOI: 10.1080/15384047.2018.1550567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Hypoxia may affect the therapeutic efficacy of transcatheter arterial embolization (TAE), which is widely used in nonsurgical hepatocellular carcinoma (HCC). Liposomal curcumin can exert anticancer effect. Our purpose is to explore the antitumor effect of liposomal curcumin on the HCC after TAE. Methods: The HepG2 cells were cultured under hypoxic condition (1% O2) and then treated with curcumin liposome. Cell viability, apoptosis and cell cycle were respectively measured by CCK-8 and a flow cytometry. The VX2 rabbits were randomly distributed into three groups: control group with saline embolization, TAE group with lipiodol embolization and curcumin liposome group with curcumin liposome and lipiodol embolization. MRI and CT perfusion scanning were performed after embolization. The hepatocyte apoptosis was measured by the terminal deoxyribonucleotidyl transferse-mediated dUTP nick-end labelling (TUNEL). The vascular endothelial growth factor (VEGF) and microvessel density (MVD) were measured by immunohistochemical. RT-PCR and Western blot were performed to examine mRNA and protein levels. Results: By regulating the apoptosis-related molecules, curcumin liposome obviously inhibited the cell viability and promoted the apoptosis in G1 phase. Curcumin liposome reduced the tumor size and alleviated neoplasia in VX2 rabbits. Curcumin liposome decreased the expressions of MVD and VEGF and increased the apoptosis of liver tissues. The levels of hypoxia-inducible factor-1α (HIF-1α) and survivin were suppressed by curcumin liposome both in hypoxic cells and liver tissues in the VX2 rabbits. Conclusion: Curcumin liposome exerted antitumor effect by regulating the proliferation- and apoptosis-related molecules. Curcumin liposome suppressed the HIF-1α and survivin levels and inhibited the angiogenesis in VX2 rabbits after TAE.
Collapse
Affiliation(s)
- Xiuming Zhang
- a Department of Radiology , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Feng Dai
- b Department of Intervention , The Second Hospital of Nanjing , Nanjing , China
| | - Jun Chen
- c Department of Intervention , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Xiaodong Xie
- a Department of Radiology , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Hanzi Xu
- d Department of Radiotherapy , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Chenguang Bai
- a Department of Radiology , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Wei Qiao
- a Department of Radiology , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| | - Wenrong Shen
- a Department of Radiology , Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital , Nanjing , China
| |
Collapse
|
21
|
Li D, Jin F, Ni T, Tao L, Wang H, Shi Q, Qian Y, Liu Y, Diao Y. Mechanism of Jinlong Capsule (JLC) in Human Esophageal Squamous Cell Carcinoma (ESCC) via the MAPK Signal Pathway. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2019.74.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
Pi C, Yuan J, Liu H, Zuo Y, Feng T, Zhan C, Wu J, Ye Y, Zhao L, Wei Y. In vitro and in vivo evaluation of curcumin loaded hollow microspheres prepared with ethyl cellulose and citric acid. Int J Biol Macromol 2018; 115:1046-1054. [PMID: 29727658 DOI: 10.1016/j.ijbiomac.2018.04.171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/31/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022]
Abstract
Curcumin (CUR) demonstrates a variety of biological activities; however, the poor oral bioavailability limits its clinical application. The objective of this study was to develop and evaluate characteristics and bioavailability of hollow microspheres loading curcumin (CUR-HPs). CUR-HPs were prepared by solvent diffusion and evaporation method. The effect of viscosity of ethyl cellulose (EC), amount of EC, citric acid (CA) and CUR on physicochemical characteristics and in vitro release profile of CUR-HPs were evaluated. Scanning electron microscopy (SEM) showed microspheres had smooth surfaces with hollow structures. The yield of CUR-HPs was (96 ± 1.80) %. The floating rate at 24 h was (89.67 ± 4.91) % and the drug loading was (3.41 ± 0.21) %. Nearly 95% of CUR was released from the HPs at 24 h. In vitro release profiles of CUR-HPs fitted the Korsmeyer et al.'s equation and indicated that CUR was released through the combination of diffusion and erosion mechanisms. The bioavailability of CUR-HPs was 12-fold higher than that of CUR. The peak time was delayed for 7.5 h and peak concentration of CUR-HPs was 3.21 times than that of free CUR. The CUR-HPs might be a promising strategy to achieve sustained release and increase oral bioavailability of CUR.
Collapse
Affiliation(s)
- Chao Pi
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Jiyuan Yuan
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Hao Liu
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Ying Zuo
- Department of General Internal Medicine, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Ting Feng
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Chenglin Zhan
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China
| | - Jun Wu
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, 307 N. Broad Street, Clinton, SC 29325, USA
| | - Yun Ye
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China; Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, No.25, Taiping Street, Luzhou, Sichuan 646000, PR China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China.
| | - Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, No. 3-5, Zhongshan Road, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
23
|
Corylin increases the sensitivity of hepatocellular carcinoma cells to chemotherapy through long noncoding RNA RAD51-AS1-mediated inhibition of DNA repair. Cell Death Dis 2018; 9:543. [PMID: 29749376 PMCID: PMC5945779 DOI: 10.1038/s41419-018-0575-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/22/2022]
Abstract
Corylin, a biologically active agent extracted from Psoralea corylifolia L. (Fabaceae), promotes bone differentiation and inhibits inflammation. Currently, few reports have addressed the biological functions that are regulated by corylin, and to date, no studies have investigated its antitumor activity. In this study, we used cell functional assays to analyze the antitumor activity of corylin in hepatocellular carcinoma (HCC). Furthermore, whole-transcriptome assays were performed to identify the downstream genes that were regulated by corylin, and gain-of-function and loss-of-function experiments were conducted to examine the regulatory roles of the above genes. We found that corylin significantly inhibited the proliferation, migration, and invasion of HCC cells and increased the toxic effects of chemotherapeutic agents against HCC cells. These properties were due to the induction of a long noncoding RNA, RAD51-AS1, which bound to RAD51 mRNA, thereby inhibiting RAD51 protein expression, thus inhibiting the DNA damage repair ability of HCC cells. Animal experiments also showed that a combination treatment with corylin significantly increased the inhibitory effects of the chemotherapeutic agent etoposide (VP16) on tumor growth. These findings indicate that corylin has strong potential as an adjuvant drug in HCC treatment and that corylin can strengthen the therapeutic efficacy of chemotherapy and radiotherapy.
Collapse
|
24
|
Masuelli L, Benvenuto M, Di Stefano E, Mattera R, Fantini M, De Feudis G, De Smaele E, Tresoldi I, Giganti MG, Modesti A, Bei R. Curcumin blocks autophagy and activates apoptosis of malignant mesothelioma cell lines and increases the survival of mice intraperitoneally transplanted with a malignant mesothelioma cell line. Oncotarget 2018; 8:34405-34422. [PMID: 28159921 PMCID: PMC5470978 DOI: 10.18632/oncotarget.14907] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/13/2016] [Indexed: 12/15/2022] Open
Abstract
Malignant mesothelioma (MM) is a primary tumor arising from the serous membranes. The resistance of MM patients to conventional therapies, and the poor patients’ survival, encouraged the identification of molecular targets for MM treatment. Curcumin (CUR) is a “multifunctional drug”. We explored the in vitro effects of CUR on cell proliferation, cell cycle regulation, pro-survival signaling pathways, apoptosis, autophagy of human (MM-B1, H-Meso-1, MM-F1), and mouse (#40a) MM cells. In addition, we evaluated the in vivo anti-tumor activities of CUR in C57BL/6 mice intraperitoneally transplanted with #40a cells forming ascites. CUR in vitro inhibited MM cells survival in a dose- and time-dependent manner and increased reactive oxygen species’intracellular production and induced DNA damage. CUR triggered autophagic flux, but the process was then blocked and was coincident with caspase 8 activation which activates apoptosis. CUR-mediated apoptosis was supported by the increase of Bax/Bcl-2 ratio, increase of p53 expression, activation of caspase 9, cleavage of PARP-1, increase of the percentage of cells in the sub G1 phase which was reduced (MM-F1 and #40a) or abolished (MM-B1 and H-Meso-1) after MM cells incubation with the apoptosis inhibitor Z-VAD-FMK. CUR treatment stimulated the phosphorylation of ERK1/2 and p38 MAPK, inhibited that of p54 JNK and AKT, increased c-Jun expression and phosphorylation and prevented NF-κB nuclear translocation. Intraperitoneal administration of CUR increased the median survival of C57BL/6 mice intraperitoneally transplanted with #40a cells and reduced the risk of developing tumors. Our findings may have important implications for the design of MM treatment using CUR.
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Enrica Di Stefano
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Rosanna Mattera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo Fantini
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Enrico De Smaele
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Ilaria Tresoldi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy.,Center for Regenerative Medicine, (CIMER), University of Rome "Tor Vergata", Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy.,Center for Regenerative Medicine, (CIMER), University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
25
|
Corylin Suppresses Hepatocellular Carcinoma Progression via the Inhibition of Epithelial-Mesenchymal Transition, Mediated by Long Noncoding RNA GAS5. Int J Mol Sci 2018; 19:ijms19020380. [PMID: 29382035 PMCID: PMC5855602 DOI: 10.3390/ijms19020380] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/13/2018] [Accepted: 01/18/2018] [Indexed: 12/16/2022] Open
Abstract
Corylin is a flavonoid extracted from the nuts of Psoralea corylifolia L. (Fabaceae), which is a widely used anti-inflammatory and anticancer herb in China. Recent studies revealed antioxidant, anti-inflammatory, and bone differentiation–promoting effects of corylin. However, there are no studies examining the anticancer activity of corylin. In this study, we used cells and animal models to examine the antitumor effects of corylin on hepatocellular carcinoma (HCC) and then studied its downstream regulatory mechanisms. The results showed that corylin significantly inhibited the proliferation, migration, and invasiveness of HCC cells and suppressed epithelial–mesenchymal transition. We found that the anti-HCC mechanism of corylin’s action lies in the upregulation of tumor suppressor long noncoding RNA growth arrest-specific transcript 5 (GAS5) and the activation of its downstream anticancer pathways. In animal experiments, we also found that corylin can significantly inhibit tumor growth without significant physiological toxicity. The above results suggest that corylin has anti-HCC effects and good potential as a clinical treatment.
Collapse
|
26
|
Early growth response 3 inhibits growth of hepatocellular carcinoma cells via upregulation of Fas ligand. Int J Oncol 2017; 50:805-814. [PMID: 28098878 DOI: 10.3892/ijo.2017.3855] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/07/2016] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignancy with aggressive biological behavior and poor prognosis. Early growth response 3 (EGR3) is a zinc finger transcription factor, and has been studied primarily in the context of neurodevelopment, autoimmunity, inflammation and angiogenesis. Accumulating evidence indicates that EGR3 is a novel suppressor gene of tumor initiation and progression in certain cancer events, but little work has been carried out in exploring the relationship between EGR3 and HCC growth. The purpose of this study was to investigate the possible effects of EGR3 on cell proliferation and apoptosis in HCC, and determine the underlying mechanisms. Here, we observed that EGR3 expression was frequently downregulated in HCC tissues and cell lines. Ectopic expression of EGR3 contributed to cell proliferation inhibition and apoptosis induction in HCC cells in vitro. Furthermore, the expression of Fas ligand (FasL) was significantly enhanced following upregulation of EGR3 in HCC cells, accompanied by an obvious increase of pro-apoptotic Bak and cell cycle inhibitor p21 expression. Based on nude mouse models, we demonstrated that ectopic expression of EGR3 markedly restricted tumor growth, and the expression of FasL was significantly increased in the xenograft tumor tissues which exhibited high EGR3 expression. We further established a co-transfection in HCC cells with EGR3 overexpression plasmid and FasL siRNA. We found that silencing of FasL gene impeded the anti-proliferative and pro-apoptotic effects, as well as the increase of Bak and p21 expression, suggesting an essential role of FasL in EGR3-mediated growth suppression in HCC cells. Collectively, in conclusion, EGR3 contributes to cell growth inhibition via upregulation of FasL in HCC.
Collapse
|
27
|
Bortel N, Armeanu-Ebinger S, Schmid E, Kirchner B, Frank J, Kocher A, Schiborr C, Warmann S, Fuchs J, Ellerkamp V. Effects of curcumin in pediatric epithelial liver tumors: inhibition of tumor growth and alpha-fetoprotein in vitro and in vivo involving the NFkappaB- and the beta-catenin pathways. Oncotarget 2016; 6:40680-91. [PMID: 26515460 PMCID: PMC4747361 DOI: 10.18632/oncotarget.5673] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/13/2015] [Indexed: 12/18/2022] Open
Abstract
In children with hepatocellular carcinoma (pHCC) the 5-year overall survival rate is poor. Effects of cytostatic therapies such as cisplatin and doxorubicin are limited due to chemoresistance and tumor relapse. In adult HCC, several antitumor properties are described for the use of curcumin. Curcumin is one of the best-investigated phytochemicals in complementary oncology without relevant side effects. Its use is limited by low bioavailability. Little is known about the influence of curcumin on pediatric epithelial hepatic malignancies. We investigated the effects of curcumin in combination with cisplatin on two pediatric epithelial liver tumor cell lines. As mechanisms of action inhibition of NFkappaB, beta-catenin, and decrease of cyclin D were identified. Using a mouse xenograft model we could show a significant decrease of alpha-fetoprotein after combination therapy of oral micellar curcumin and cisplatin. Significant concentrations of curcuminoids were found in blood samples, organ lysates, and tumor tissue after oral micellar curcumin administration. Micellar curcumin in combination with cisplatin can be a promising strategy for treatment of pediatric HCC.
Collapse
Affiliation(s)
- Nicola Bortel
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Sorin Armeanu-Ebinger
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Evi Schmid
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Bettina Kirchner
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Division of Biofunctionality and Safety of Food, D-70599 Stuttgart, Germany
| | - Alexa Kocher
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Division of Biofunctionality and Safety of Food, D-70599 Stuttgart, Germany
| | - Christina Schiborr
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Division of Biofunctionality and Safety of Food, D-70599 Stuttgart, Germany
| | - Steven Warmann
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| | - Verena Ellerkamp
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tuebingen, D-72076 Tuebingen, Germany
| |
Collapse
|
28
|
Montazeri M, Sadeghizadeh M, Pilehvar-Soltanahmadi Y, Zarghami F, Khodi S, Mohaghegh M, Sadeghzadeh H, Zarghami N. Dendrosomal curcumin nanoformulation modulate apoptosis-related genes and protein expression in hepatocarcinoma cell lines. Int J Pharm 2016; 509:244-254. [PMID: 27234697 DOI: 10.1016/j.ijpharm.2016.05.039] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/16/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022]
Abstract
The side-effects observed in conventional therapies have made them unpromising in curing Hepatocellular carcinoma; therefore, developing novel treatments can be an overwhelming significance. One of such novel agents is curcumin which can induce apoptosis in various cancerous cells, however, its poor solubility is restricted its application. To overcome this issue, this paper employed dendrosomal curcumin (DNC) was employed to in prevent hepatocarcinoma in both RNA and protein levels. Hepatocarcinoma cells, p53 wild-type HepG2 and p53 mutant Huh7, were treated with DNC and investigated for toxicity study using MTT assay. Cell cycle distribution and apoptosis were analyzed using Flow-cytometry and Annexin-V-FLUOS/PI staining. Real-time PCR and Western blot were employed to analyze p53, BAX, Bcl-2, p21 and Noxa in DNC-treated cells. DNC inhibited the growth in the form of time-dependent manner, while the carrier alone was not toxic to the cell. Flow-cytometry data showed the constant concentration of 20μM DNC during the time significantly increases cell population in SubG1 phase. Annexin-V-PI test showed curcumin-induced apoptosis was enhanced in Huh7 as well as HepG2, compared to untreated cells. Followed by treatment, mRNA expression of p21, BAX, and Noxa increased, while the expression of Bcl-2 decreased, and unlike HepG2, Huh7 showed down-regulation of p53. In summary, DNC-treated hepatocellular carcinoma cells undergo apoptosis by changing the expression of genes involved in the apoptosis and proliferation processes. These findings suggest that DNC, as a plant-originated therapeutic agent, could be applied in cancer treatment.
Collapse
Affiliation(s)
- Maryam Montazeri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Yones Pilehvar-Soltanahmadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faraz Zarghami
- Imam Reza Teaching Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Khodi
- Department of Medical Genetic, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mina Mohaghegh
- Department of Molecular Biology and Biotechnology, University of Aix-Marseille, Marseille, France
| | - Hadi Sadeghzadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Hu B, An HM, Wang SS, Chen JJ, Xu L. Preventive and Therapeutic Effects of Chinese Herbal Compounds against Hepatocellular Carcinoma. Molecules 2016; 21:142. [PMID: 26828466 PMCID: PMC6274246 DOI: 10.3390/molecules21020142] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 12/14/2022] Open
Abstract
Traditional Chinese Medicines, unique biomedical and pharmaceutical resources, have been widely used for hepatocellular carcinoma (HCC) prevention and treatment. Accumulated Chinese herb-derived compounds with significant anti-cancer effects against HCC have been identified. Chinese herbal compounds are effective in preventing carcinogenesis, inhibiting cell proliferation, arresting cell cycle, inducing apoptosis, autophagy, cell senescence and anoikis, inhibiting epithelial-mesenchymal transition, metastasis and angiogenesis, regulating immune function, reversing drug resistance and enhancing the effects of chemotherapy in HCC. This paper comprehensively reviews these compounds and their effects on HCC. Finally, the perspectives and rational application of herbal compounds for HCC management are discussed.
Collapse
Affiliation(s)
- Bing Hu
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hong-Mei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 202032, China.
| | - Shuang-Shuang Wang
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jin-Jun Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, The Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China.
| | - Ling Xu
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
30
|
Ghosh S, Banerjee S, Sil PC. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: A recent update. Food Chem Toxicol 2015; 83:111-124. [PMID: 26066364 DOI: 10.1016/j.fct.2015.05.022] [Citation(s) in RCA: 327] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 02/06/2023]
Abstract
The concept of using phytochemicals has ushered in a new revolution in pharmaceuticals. Naturally occurring polyphenols (like curcumin, morin, resveratrol, etc.) have gained importance because of their minimal side effects, low cost and abundance. Curcumin (diferuloylmethane) is a component of turmeric isolated from the rhizome of Curcuma longa. Research for more than two decades has revealed the pleiotropic nature of the biological effects of this molecule. More than 7000 published articles have shed light on the various aspects of curcumin including its antioxidant, hypoglycemic, anti-inflammatory and anti-cancer activities. Apart from these well-known activities, this natural polyphenolic compound also exerts its beneficial effects by modulating different signalling molecules including transcription factors, chemokines, cytokines, tumour suppressor genes, adhesion molecules, microRNAs, etc. Oxidative stress and inflammation play a pivotal role in various diseases like diabetes, cancer, arthritis, Alzheimer's disease and cardiovascular diseases. Curcumin, therefore, could be a therapeutic option for the treatment of these diseases, provided limitations in its oral bioavailability can be overcome. The current review provides an updated overview of the metabolism and mechanism of action of curcumin in various organ pathophysiologies. The review also discusses the potential for multifunctional therapeutic application of curcumin and its recent progress in clinical biology.
Collapse
Affiliation(s)
- Shatadal Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India
| | - Sharmistha Banerjee
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India.
| |
Collapse
|
31
|
Wang HB, Ma XQ. β, β-Dimethylacrylshikonin induces mitochondria-dependent apoptosis of human lung adenocarcinoma cells in vitro via p38 pathway activation. Acta Pharmacol Sin 2015; 36:131-8. [PMID: 25434989 DOI: 10.1038/aps.2014.108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/26/2014] [Indexed: 11/09/2022]
Abstract
AIM β, β-Dimethylacrylshikonin (DMAS) is an anticancer compound extracted from the roots of Lithospermum erythrorhizon. In the present study, we investigated the effects of DMAS on human lung adenocarcinoma cells in vitro and explored the mechanisms of its anti-cancer action. METHODS Human lung adenocarcinoma A549 cells were tested. Cell viability was assessed using an MTT assay, and cell apoptosis was evaluated with flow cytometry and DAPI staining. The expression of the related proteins was detected using Western blotting. The mitochondrial membrane potential was measured using a JC-1 kit, and subcellular distribution of cytochrome c was analyzed using immunofluorescence staining. RESULTS Treatment of A549 cells with DMAS suppressed the cell viability in dose- and time-dependent manners (the IC50 value was 14.22 and 10.61 μmol/L, respectively, at 24 and 48 h). DMAS (7.5, 10, and 15 μmol/L) dose-dependently induced apoptosis, down-regulated cIAP-2 and XIAP expression, and up-regulated Bax and Bak expression in the cells. Furthermore, DMAS resulted in loss of mitochondrial membrane potential and release of cytochrome c in the cells, and activated caspase-9, caspase-8, and caspase-3, and subsequently cleaved PARP, which was abolished by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor. DMAS induced sustained p38 phosphorylation in the cells, while pretreatment with SB203580, a specific p38 inhibitor, blocked DMAS-induced p38 activation and apoptosis. CONCLUSION DMAS inhibits the growth of human lung adenocarcinoma A549 cells in vitro via activation of p38 signaling pathway.
Collapse
|
32
|
Tuorkey MJ. Curcumin a potent cancer preventive agent: Mechanisms of cancer cell killing. Interv Med Appl Sci 2014; 6:139-46. [PMID: 25598986 DOI: 10.1556/imas.6.2014.4.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 06/29/2014] [Accepted: 08/01/2014] [Indexed: 12/13/2022] Open
Abstract
There is no doubt that diet could effectively improve health and halt cancers. Dietary phytochemical compounds and their derivatives represent a cornucopia of effectively anticancer compounds. This review discusses existing data on the anticancer activities of curcumin, and then offers possible explanations for and mechanisms of its cancer-preventive action. This review also offers insights into the molecular mechanism and targets through which curcumin modulates cell cycle, apoptotic signals, anti-apoptotic proteins, miRNAs, Wnt/beta-catenin signaling, protein kinases, nuclear factor-κB, proteasome activation, epigenetic regulation including DNA methylation and histone modification. Finally, this review provides explanations for how curcumin reverses the multi-drug resistance (MDR) of cancer cells.
Collapse
|
33
|
Shang C, Guo Y, Hong Y, Liu YH, Xue YX. MiR-21 up-regulation mediates glioblastoma cancer stem cells apoptosis and proliferation by targeting FASLG. Mol Biol Rep 2014; 42:721-7. [PMID: 25394756 DOI: 10.1007/s11033-014-3820-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 11/08/2014] [Indexed: 11/30/2022]
Abstract
To investigate whether miR-21 can affect the apoptosis and proliferation of glioblastoma cancer stem cells (GSCs) from down-regulating FASLG. The expression of miRNA-21 was detected by quantitative real-time PCR in normal brain tissue and glioblastoma samples, and the changes of miRNA-21 expression between GSCs and non-GSCs were also detected. The apoptosis and proliferation ability of miR-21 in GSCs were analyzed by MTT and flow cytometry assay after anti-miR-21 transfection. For the regulation mechanism analysis of miR-21, TargetScan, PicTar and microRNA were selected to predict some potential target genes of miR-21. The predicted gene was identified to be the direct and specific target gene of miR-21 by luciferase activities assay and western blot. RNA interference technology was used to confirm the apoptosis and proliferation effects of miR-21 were directly induced by FASLG. The expression of miR-21 increased significantly in glioblastoma contrast to normal brain tissue, and miR-21 up-regulated in GSCs remarkably. The proliferation of GSCs cell could be inhibited with high-expression of miR-21 and this effect could be restored by miR-21 knocked down. Mechanism analysis revealed that FASLG was a specific and direct target gene of miR-21. The advanced effects of anti-miR-21 on GSCs apoptosis and proliferation were mediated by expression of silenced FASLG. In summary, aberrantly expressed miR-21 regulates GSCs apoptosis and proliferation partly through directly down-regulating FASLG protein expression in GSCs and this might offer a new potential therapeutic stratagem for glioblastoma.
Collapse
Affiliation(s)
- Chao Shang
- Department of Neurobiology, China Medical University, No. 92 Beier Road, Heping District, Shenyang, 110001, People's Republic of China
| | | | | | | | | |
Collapse
|
34
|
Liang T, Zhang X, Xue W, Zhao S, Zhang X, Pei J. Curcumin induced human gastric cancer BGC-823 cells apoptosis by ROS-mediated ASK1-MKK4-JNK stress signaling pathway. Int J Mol Sci 2014; 15:15754-65. [PMID: 25198898 PMCID: PMC4200840 DOI: 10.3390/ijms150915754] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/26/2014] [Accepted: 09/03/2014] [Indexed: 11/16/2022] Open
Abstract
The signaling mediated by stress-activated MAP kinases (MAPK), c-Jun N-terminal kinase (JNK) has well-established importance in cancer. In the present report, we investigated the effects of curcumin on the signaling pathway in human gastric cancer BGC-823 cells. Curcumin induced reactive oxygen species (ROS) production and BGC-823 cells apoptosis. Inhibition of ROS generation by antioxidant (NAC or Trion) significantly prevented curcumin-mediated apoptosis. Notably, we observed that curcumin activated ASK1, a MAPKKK that is oxidative stress sensitive and responsible to phosphorylation of JNK via triggering cascades, up-regulated an upstream effector of the JNK, MKK4, and phosphorylated JNK protein expression in BGC-823 cells. However, curcumin induced ASK1-MKK4-JNK signaling was attenuated by NAC. All the findings confirm the possibility that oxidative stress-activated ASK1-MKK4-JNK signaling cascade promotes the apoptotic response in curcumin-treated BGC-823 cells.
Collapse
Affiliation(s)
- Tao Liang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiaojian Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Wenhua Xue
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Songfeng Zhao
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiang Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jianying Pei
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
35
|
Chaudhuri J, Chowdhury AA, Biswas N, Manna A, Chatterjee S, Mukherjee T, Chaudhuri U, Jaisankar P, Bandyopadhyay S. Superoxide activates mTOR-eIF4E-Bax route to induce enhanced apoptosis in leukemic cells. Apoptosis 2014; 19:135-48. [PMID: 24052408 DOI: 10.1007/s10495-013-0904-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a central kinase that regulates cell survival, proliferation and translation. Reactive oxygen species (ROS) are second messengers with potential in manipulating cellular signaling. Here we report that two ROS generating phytochemicals, hydroxychavicol and curcumin synergize in leukemic cells in inducing enhanced apoptosis by independently activating both mitogen activated protein kinase (MAPK) (JNK and P(38)) and mTOR pathways. Low level transient ROS generated after co-treatment with these phytochemicals led to activation of these two pathways. Both mTOR and MAPK pathways played important roles in co-treatment-induced apoptosis, by knocking down either mTOR or MAPKs inhibited apoptosis. Activation of mTOR, as evident from phosphorylation of its downstream effector eukaryotic translation initiation factor 4E-binding protein 1, led to release of eukaryotic translation initiation factor 4E (eIF4E) which was subsequently phosphorylated by JNK leading to translation of pro-apoptotic proteins Bax and Bad without affecting the expression of anti-apoptotic protein Bcl-xl. Our data suggest that mTOR and MAPK pathways converge at eIF4E in co-treatment-induced enhanced apoptosis and provide mechanistic insight for the role of mTOR activation in apoptosis.
Collapse
Affiliation(s)
- Jaydeep Chaudhuri
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen J, Li L, Su J, Li B, Chen T, Wong YS. Synergistic apoptosis-inducing effects on A375 human melanoma cells of natural borneol and curcumin. PLoS One 2014; 9:e101277. [PMID: 24971451 PMCID: PMC4074168 DOI: 10.1371/journal.pone.0101277] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 06/04/2014] [Indexed: 12/12/2022] Open
Abstract
This study was to investigate the synergistic effect of NB/Cur on growth and apoptosis in A375 human melanoma cell line by MTT assay, flow cytometry and Western blotting. Our results demonstrated that NB effectively synergized with Cur to enhance its antiproliferative activity on A375 human melanoma cells by induction of apoptosis, as evidenced by an increase in sub-G1 cell population, DNA fragmentation, PARP cleavage and caspase activation. Further mechanistic studies by Western blotting showed that after treatment of the cells with NB/Cur, up-regulation of the expression level of phosphorylated JNK and down-regulation of the expression level of phosphorylated ERK and Akt contributed to A375 cells apoptosis. Moreover, NB also potentiated Cur to trigger intracellular ROS overproduction and the DNA damage with up-regulation of the expression level of phosphorylated ATM, phosphorylated Brca1 and phosphorylated p53. The results indicate the combinational application potential of NB and Cur in treatments of cancers.
Collapse
Affiliation(s)
- Jianping Chen
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, China
| | - Lin Li
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, China
| | - Jianyu Su
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, China
- Guangdong Hua Qing Yuan Biological Technology Co., Ltd., Meizhou, China
- * E-mail: (JS); (TC)
| | - Bing Li
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, China
- Guangdong Hua Qing Yuan Biological Technology Co., Ltd., Meizhou, China
- * E-mail: (JS); (TC)
| | - Yum-Shing Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
37
|
Wu J, Tang Q, Zhao S, Zheng F, Wu Y, Tang G, Hahn SS. Extracellular signal-regulated kinase signaling-mediated induction and interaction of FOXO3a and p53 contribute to the inhibition of nasopharyngeal carcinoma cell growth by curcumin. Int J Oncol 2014; 45:95-103. [PMID: 24806432 PMCID: PMC4079154 DOI: 10.3892/ijo.2014.2420] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/02/2014] [Indexed: 01/03/2023] Open
Abstract
Curcumin, one of the main bioactive components extracted from a traditional Chinese medicinal herb, exhibits potent anticancer activity against many types of cancer cells including nasopharyngeal carcinoma (NPC). However, the detailed molecular mechanism underlying this is not clearly understood. In this study, we showed that curcumin significantly inhibited the growth of NPC cells in a dose-and time-dependent manner as determined by MTT assays, while increasing apoptosis was also observed as measured by flow cytometry for the FITC-Annexin V and propidium iodide (PI) label and Hoechst 33258 staining. To further explore the potential mechanism, we showed that curcumin increased the phosphorylation of ERK1/2 but not p38 MAPK in a time-dependent manner, and induced protein expression of the tumor suppressors FOXO3a and p53 in a dose-dependent manner, which were not observed in the presence of PD98059, an inhibitor of ERK1/2. Furthermore, silencing of FOXO3a and p53 genes by siRNAs overcame the inhibitory effect of curcumin on cell proliferation. Silencing or blockade of p53 using siRNA or chemical inhibitor abrogated the effect of curcumin on expression of FOXO3a protein; silencing or overexpression of FOXO3a had no further effect on curcumin-induced p53 protein expression. Furthermore, blockade of ERK1/2 and exogenous expression of FOXO3a restored the effect of curcumin on growth of cells. Together, our studies show that curcumin inhibits growth and induces apoptosis of NPC cells through ERK1/2-mediated increase in the protein expression and interaction of p53 and FOXO3a. p53 is upstream of FOXO3a, which form a regulatory loop that mediates the effect of curcumin. This study unveils a new mechanism by which curcumin inhibits the proliferation and induces apoptosis of human NPC cells.
Collapse
Affiliation(s)
- Jingjing Wu
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Qin Tang
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Shunyu Zhao
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Fang Zheng
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Yan Wu
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Ge Tang
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Swei Sunny Hahn
- Laboratory of Tumor Molecular Biology and Targeted Therapies of Chinese Medicine, University of Guangzhou Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, P.R. China
| |
Collapse
|
38
|
Nabavi SF, Daglia M, Moghaddam AH, Habtemariam S, Nabavi SM. Curcumin and Liver Disease: from Chemistry to Medicine. Compr Rev Food Sci Food Saf 2013; 13:62-77. [DOI: 10.1111/1541-4337.12047] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/23/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Seyed Fazel Nabavi
- Applied Biotechnology Research Center; Baqiyatallah Univ. of Medical Sciences; Tehran Iran
| | - Maria Daglia
- Dept. of Drug Sciences; Univ. of Pavia, Medicinal Chemistry and Pharmaceutical Technology Section; via Taramelli 12 27100 Pavia Italy
| | - Akbar Hajizadeh Moghaddam
- Amol Univ. of Special Modern Technologies; Amol Iran
- Dept. of Biology; Faculty of basic science; Univ. of Mazandaran; Babolsar Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories; Medway School of Science, Univ. of Greenwich; Central Ave. Chatham-Maritime Kent ME4 4TB U.K
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center; Baqiyatallah Univ. of Medical Sciences; Tehran Iran
| |
Collapse
|
39
|
Kim JS, Park MR, Lee SY, Kim DK, Moon SM, Kim CS, Cho SS, Yoon G, Im HJ, You JS, Oh JS, Kim SG. Licochalcone A induces apoptosis in KB human oral cancer cells via a caspase-dependent FasL signaling pathway. Oncol Rep 2013; 31:755-62. [PMID: 24337492 PMCID: PMC3983909 DOI: 10.3892/or.2013.2929] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/25/2013] [Indexed: 12/11/2022] Open
Abstract
Licochalcone A (Lico-A) is a natural phenol licorice compound with multiple bioactivities, including anti-inflammatory, anti-microbial, anti-fungal and osteogenesis-inducing properties. In the present study, we investigated the Lico-A-induced apoptotic effects and examined the associated apoptosis pathway in KB human oral cancer cells. Lico-A decreased the number of viable KB oral cancer cells. However, Lico-A did not have an effect on primary normal human oral keratinocytes. In addition, the IC50 value of Lico-A was determined to be ~50 μM following dose-dependent stimulation. KB oral cancer cells stimulated with Lico-A for 24 h showed chromatin condensation by DAPI staining, genomic DNA fragmentation by agarose gel electrophoresis and a gradually increased apoptotic cell population by FACS analysis. These data suggest that Lico-A induces apoptosis in KB oral cancer cells. Additionally, Lico-A-induced apoptosis in KB oral cancer cells was mediated by the expression of factor associated suicide ligand (FasL) and activated caspase-8 and −3 and poly(ADP-ribose) polymerase (PARP). Furthermore, in the KB oral cancer cells co-stimulation with a caspase inhibitor (Z-VAD-fmk) and Lico-A significantly abolished the apoptotic phenomena. Our findings demonstrated that Lico-A-induced apoptosis in KB oral cancer cells involves the extrinsic apoptotic signaling pathway, which involves a caspase-dependent FasL-mediated death receptor pathway. Our data suggest that Lico-A be developed as a chemotherapeutic agent for the management of oral cancer.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Mi-Ra Park
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Sook-Young Lee
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Do Kyoung Kim
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Sung-Min Moon
- Department of Oral Biochemistry, Chosun University, Gwangju 501-759, Republic of Korea
| | - Chun Sung Kim
- Department of Oral Biochemistry, Chosun University, Gwangju 501-759, Republic of Korea
| | - Seung Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Jeonnam 535-729, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Jeonnam 535-729, Republic of Korea
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jae-Seek You
- Department of Oral and Maxillofacial Surgery, Chosun University, Gwangju 501-759, Republic of Korea
| | - Ji-Su Oh
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Su-Gwan Kim
- Regional Innovation Center for Dental Science and Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| |
Collapse
|
40
|
Liu J, Tang SX. Molecular mechanisms behind anti-cancer effects of curcumin in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:3678-3682. [DOI: 10.11569/wcjd.v21.i33.3678] [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: 02/06/2023] Open
Abstract
Primary liver cancer, also known as hepatocellular carcinoma (HCC), is one of the most lethal cancers worldwide. Most HCC cases are reported in the developing countries in Asia and Africa. Viral hepatitis, alcoholism as well as dietary carcinogens, such as aflatoxins and nitrosoamines, contribute to HCC. Surgical resection is the best treatment for early HCC, and Chinese medicine is one of the main measures for the treatment of advanced HCC. Numerous studies show that curcumin has anti-cancer, anti-oxidant and anti-inflammatory properties. This article will discuss the mechanisms behind anti-cancer effects of curcumin in HCC.
Collapse
|
41
|
Li JK, Liang HL, Li Z, Gu CH, Yi DH, Pei JM. Pigment epithelium-derived factor promotes Fas-induced cardiomyocyte apoptosis via its receptor phospholipase A2. Life Sci 2013; 99:18-23. [PMID: 23892196 DOI: 10.1016/j.lfs.2013.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/08/2013] [Accepted: 07/12/2013] [Indexed: 12/19/2022]
Abstract
AIMS Cardiovascular diseases cause significant morbidity and mortality worldwide. Recently, our research team demonstrated that a multifunctional cytokine, pigment epithelium-derived factor (PEDF), plays a critical role in regulating myocardial infarction. However, few researchers have studied the molecular mechanisms by which PEDF and its receptors influence the pathophysiology of cardiovascular disease. We tested the hypothesis that PEDF affects cardiomyocyte apoptosis under hypoxic conditions and determined the role that its receptors phospholipase A2 (PLA2) and laminin receptor play in this process. MAIN METHODS Cardiomyocytes were isolated from neonatal mice and treated with PEDF under normoxic and hypoxic conditions; then, apoptosis was assessed using Annexin V/PI staining and flow cytometry. Western blotting and immunofluorescence staining were used to detect PEDF receptor expression, and siRNA knockdown of PEDF receptors was performed to determine which receptor was involved in mediating cardiomyocyte apoptosis. KEY FINDINGS Our results demonstrated that PEDF increased cardiomyocyte apoptosis during hypoxia via Fas and that PEDF receptors were expressed on cardiomyocyte cell membranes. Furthermore, siRNA experiments indicated that the PEDF receptor PLA2 was responsible for inducing cardiomyocyte apoptosis via the Fas pathway. SIGNIFICANCE PEDF promoted Fas-induced cardiomyocyte apoptosis via its receptor PLA2.
Collapse
Affiliation(s)
- Ji-ke Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China
| | - Hong-liang Liang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China
| | - Zhi Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China
| | - Chun-hu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China
| | - Ding-hua Yi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China.
| | - Jian-ming Pei
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No. 172 West Changle Rd, Xi'an 710032, China; Department of Physiology, Fourth Military Medical University, No. 169 West Changle Rd, Xi'an, 710032, China.
| |
Collapse
|
42
|
Liu TY, Tan ZJ, Jiang L, Gu JF, Wu XS, Cao Y, Li ML, Wu KJ, Liu YB. Curcumin induces apoptosis in gallbladder carcinoma cell line GBC-SD cells. Cancer Cell Int 2013; 13:64. [PMID: 23802572 PMCID: PMC3733655 DOI: 10.1186/1475-2867-13-64] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 06/23/2013] [Indexed: 12/15/2022] Open
Abstract
Background Gallbladder carcinoma is a malignant tumor with a very low 5-year survival rate because of the difficulty with its early diagnosis and the very poor prognosis of the advanced cancer state. The aims of this study were to determine whether curcumin could induce the apoptosis of a gallbladder carcinoma cell line, GBC-SD, and to clarify its related mechanism. Methods First, the anti-proliferative activities of curcumin-treated and untreated GBC-SD cells were determined using the MTT and colony formation assays. Then, the early apoptosis of cells was detected by the annexin V/propidium iodide double-staining assay and Hoechst 33342 staining assay. Detection of mitochondrial membrane potential was used to validate the ability of curcumin on inducing apoptosis in GBC-SD cells. Cell cycle changes were detected by flow cytometric analysis. Finally, the expressions of the apoptosis-related proteins or genes caspase-3, PARP, Bcl-2, and Bax were analyzed by western blot and quantitative real time PCR assay. Statistical analyses were performed using the Student’s t-test for comparison of the results obtained from cells with or without curcumin treatment. Results The MTT assay revealed that curcumin had induced a dose- and a time-dependent decrease in cell viability. Colony counting indicated that curcumin had induced a dose-dependent decrease in the colony formation ability in GBC-SD cells. Cells treated with curcumin were arrested at the S phase, according to the flow cytometric analysis. A significant induction of both the early and late phases of apoptosis was shown by the annexin V-FITC and PI staining. Morphological changes in apoptotic cells were also found by the Hoechst 33342 staining. After treatment with curcumin fluorescence shifted from red to green as ΔΨm decreased. Furthermore, western blot and quantitative real time PCR assays demonstrated that the curcumin induced apoptosis in GBC-SD cells by regulating the ratio of Bcl-2/Bax and activating the expression of cleaved caspase-3. Conclusions Taken together, the results indicate that curcumin may be a potential agent for the treatment of gallbladder cancer.
Collapse
Affiliation(s)
- Tian-Yu Liu
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, No, 1665 Kongjiang Road, Shanghai 200092, China.
| | | | | | | | | | | | | | | | | |
Collapse
|