1
|
Du NH, Ngoc TTB, Cang HQ, Luyen NTT, Thuoc TL, Le Quan T, Thao DTP. KTt-45, a T-type calcium channel blocker, acts as an anticancer agent by inducing apoptosis on HeLa cervical cancer cell line. Sci Rep 2023; 13:22092. [PMID: 38086845 PMCID: PMC10716508 DOI: 10.1038/s41598-023-47199-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
The abnormal expression in the T-type calcium channels is involved in various cancer types, thus inhibiting T-type calcium channels is one of approaches in cancer treatment. The fact that KTt-45 acted as a T-type calcium channel inhibitor as well as a pain-relief agent prompts us to address if KTt-45 plays any role against cancer cells. The results showed that KTt-45 caused cytotoxic effects towards HeLa cervical, Raji lymphoma, MCF-7 breast cancer, and A549 lung cancer cell lines with IC50 values less than 100 μM, in which highly selective toxicity was against HeLa cells (IC50 = 37.4 μM, SI > 3.2). Strikingly, the KTt-45 induced an accumulation of cytoplasmic vacuoles after 48 h treatment and mitochondrial-dependent apoptosis activation as evidenced by morphological features, chromatin condensation, nuclear fragmentation, and significant activation of caspase-9 as well as caspase-3. In conclusion, KTt-45 could inhibit cell growth and trigger mitochondrial-dependent apoptosis in HeLa cervical cancer cells. The results, taken together, strongly demonstrated that KTt-45 is a potential agent for further study on anticancer drug development which not only targets cancer cells but also helps to relieve neuropathic pain in cancer patients.
Collapse
Affiliation(s)
- Nguyen Huy Du
- Department of Molecular and Environmental Biotechnology, Faculty of Biology and Biotechnology, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Laboratory of Cancer Research, VNU-HCM, University of Science, Duong so 4, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Central Laboratory of Analysis, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
| | - Truong Thi Bich Ngoc
- Department of Molecular and Environmental Biotechnology, Faculty of Biology and Biotechnology, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Laboratory of Cancer Research, VNU-HCM, University of Science, Duong so 4, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
| | - Huynh Qui Cang
- Laboratory of Cancer Research, VNU-HCM, University of Science, Duong so 4, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
| | - Nguyen Thi Thuy Luyen
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Central Laboratory of Analysis, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Department of Hydro-Geology-Engineering Geology and Environmental Geology, Faculty of Geology, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Department of Medicinal Chemistry, Faculty of Chemistry, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
| | - Tran Linh Thuoc
- Laboratory of Cancer Research, VNU-HCM, University of Science, Duong so 4, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
| | - Tran Le Quan
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam
- Central Laboratory of Analysis, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Department of Hydro-Geology-Engineering Geology and Environmental Geology, Faculty of Geology, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
- Department of Medicinal Chemistry, Faculty of Chemistry, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam
| | - Dang Thi Phuong Thao
- Department of Molecular and Environmental Biotechnology, Faculty of Biology and Biotechnology, VNU-HCM, University of Science, 227 Nguyen Van Cu, Ho Chi Minh City, 700000, Vietnam.
- Laboratory of Cancer Research, VNU-HCM, University of Science, Duong so 4, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University, Ho Chi Minh City, Vo Truong Toan, Linh Trung, Thu Duc, Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
2
|
Zhong J, Fang L, Chen R, Xu J, Guo D, Guo C, Guo C, Chen J, Chen C, Wang X. Polysaccharides from sporoderm-removed spores of Ganoderma lucidum induce apoptosis in human gastric cancer cells via disruption of autophagic flux. Oncol Lett 2021; 21:425. [PMID: 33850566 PMCID: PMC8025153 DOI: 10.3892/ol.2021.12686] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/24/2022] Open
Abstract
The sporoderm-broken spores of Ganoderma lucidum (G. lucidum) polysaccharide (BSGLP) have been demonstrated to inhibit carcinogenesis in several types of cancer. However, to the best of our knowledge, the anticancer effects of polysaccharides extracted from the newly developed sporoderm-removed spores of G. lucidum (RSGLP) have not been assessed. The present study first compared the anticancer effects of RSGLP and BSGLP in three gastric cancer cell lines and it was found that RSGLP was more potent than BSGLP in decreasing gastric cancer cell viability. RSGLP significantly induced apoptosis in AGS cells, accompanied by downregulation of Bcl-2 and pro-caspase-3 expression levels, and upregulation of cleaved-PARP. Furthermore, RSGLP increased LC3-II and p62 expression, indicative of induction of autophagy and disruption of autophagic flux in AGS cells. These results were further verified by combined treatment of AGS cells with the late-stage autophagy inhibitor chloroquine, or early-stage autophagy inducer rapamycin. Adenoviral transfection with mRFP-GFP-LC3 further confirmed that autophagic flux was inhibited by RSGLP in AGS cells. Finally, the present study demonstrated that the RSGLP-induced autophagy and disruption of autophagic flux disruption was, at least in part, responsible for RSGLP-induced apoptosis in AGS cells. The results of the present study demonstrated for the first time that RSGLP is more effective than BSGLP in inhibiting gastric cancer cell viability, and RSGLP may serve as a promising autophagy inhibitor in the management of gastric cancer.
Collapse
Affiliation(s)
- Jiayi Zhong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China.,Department of Pharmacy, Wenling Maternal and Child Health Care Hospital, Taizhou, Zhejiang 317500, P.R. China
| | - Liu Fang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Rong Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jing Xu
- Zhejiang Engineering Research Center of Rare Medicinal Plants, Wuyi, Zhejiang 321200, P.R. China
| | - Dandan Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Chengjie Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Cuiling Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jiajun Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Chaojie Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xingya Wang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
3
|
Zhang B, Liu L. Autophagy is a double-edged sword in the therapy of colorectal cancer. Oncol Lett 2021; 21:378. [PMID: 33777202 PMCID: PMC7988732 DOI: 10.3892/ol.2021.12639] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-associated mortality worldwide. The limitations of colorectal cancer treatment include various types of multidrug resistance and the contingent damage to neighboring normal cells caused by chemotherapy. Macroautophagy/autophagy and apoptosis are essential mechanisms involved in cancer cell regulation of chemotherapy. Autophagy can either cause cancer cell death or promote tumor survival during colorectal cancer. Given that autophagy is involved in chemotherapy of colorectal cancer, an improved insight into the potential interactions between apoptosis and autophagy is crucial. The present review aimed to summarize the involvement of autophagy in the regulation of colorectal cancer and its association with chemotherapy. Furthermore, the role of natural product extraction, novel chemicals and small molecules, as well as radiation, which induce autophagy in colorectal cancer cells, were reviewed. Finally, the present review aimed to provide an outlook for the regulation of autophagy as a novel approach to the treatment of cancer, particularly chemotherapy-resistant colorectal cancer.
Collapse
Affiliation(s)
- Bo Zhang
- Medical Laboratory for Radiation Research, Beijing Institute for Occupational Disease Prevention and Treatment, Beijing 100093, P.R. China.,College of Food Science and Engineering, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Lantao Liu
- Medical Laboratory for Radiation Research, Beijing Institute for Occupational Disease Prevention and Treatment, Beijing 100093, P.R. China
| |
Collapse
|
4
|
Kim MJ, Kang YJ, Sung B, Jang JY, Ahn YR, Oh HJ, Choi H, Choi I, Im E, Moon HR, Chung HY, Kim ND. Novel SIRT Inhibitor, MHY2256, Induces Cell Cycle Arrest, Apoptosis, and Autophagic Cell Death in HCT116 Human Colorectal Cancer Cells. Biomol Ther (Seoul) 2020; 28:561-568. [PMID: 33073770 PMCID: PMC7585637 DOI: 10.4062/biomolther.2020.153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
We examined the anticancer effects of a novel sirtuin inhibitor, MHY2256, on HCT116 human colorectal cancer cells to investigate its underlying molecular mechanisms. MHY2256 significantly suppressed the activity of sirtuin 1 and expression levels of sirtuin 1/2 and stimulated acetylation of forkhead box O1, which is a target protein of sirtuin 1. Treatment with MHY2256 inhibited the growth of the HCT116 (TP53 wild-type), HT-29 (TP53 mutant), and DLD-1 (TP53 mutant) human colorectal cancer cell lines. In addition, MHY2256 induced G0/G1 phase arrest of the cell cycle progression, which was accompanied by the reduction of cyclin D1 and cyclin E and the decrease of cyclin-dependent kinase 2, cyclin-dependent kinase 4, cyclin-dependent kinase 6, phosphorylated retinoblastoma protein, and E2F transcription factor 1. Apoptosis induction was shown by DNA fragmentation and increase in late apoptosis, which were detected using flow cytometric analysis. MHY2256 downregulated expression levels of procaspase-8, -9, and -3 and led to subsequent poly(ADP-ribose) polymerase cleavage. MHY2256-induced apoptosis was involved in the activation of caspase-8, -9, and -3 and was prevented by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor. Furthermore, the autophagic effects of MHY2256 were observed as cytoplasmic vacuolation, green fluorescent protein-light-chain 3 punctate dots, accumulation of acidic vesicular organelles, and upregulated expression level of light-chain 3-II. Taken together, these results suggest that MHY2256 could be a potential novel sirtuin inhibitor for the chemoprevention or treatment of colorectal cancer or both.
Collapse
Affiliation(s)
- Min Jeong Kim
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Young Jung Kang
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Bokyung Sung
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Jung Yoon Jang
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Yu Ra Ahn
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Hye Jin Oh
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Heejeong Choi
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Inkyu Choi
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Eunok Im
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Hyung Ryong Moon
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Hae Young Chung
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Nam Deuk Kim
- Division of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
5
|
Li K, Hao K, Zhang Y, Xu A, Wang Q, Du Y, Wu L, Chen B, Zhang W, Wang Z. C21 Fraction Refined from Marsdenia tenacissima-Induced Apoptosis is Enhanced by Suppression of Autophagy in Human Gastric Cell Lines. ACS OMEGA 2020; 5:25156-25163. [PMID: 33043194 PMCID: PMC7542599 DOI: 10.1021/acsomega.0c02748] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
C21 steroidal glycosides have been extensively reported for treating several types of cancer and are widely found in Marsdenia tenacissima. In this study, a C21 fraction was synthesized from M. tenacissima, and its anti-cancer potency was assessed against in vitro gastric cell lines BGC-823, SGC-7901, and AGS. Significant growth inhibition and cell cycle arrest were observed in C21 fraction-treated gastric cancer cells. The results of apoptotic staining techniques in C21 fraction-treated gastric cells were confirmed with excess reactive oxygen species generation. Moreover, SOD and H2O2 levels were increased by C21 fraction, especially when combined with chloroquine (CQ). The apoptotic inducing potential of C21 fraction was also evidenced by upregulation of proapoptotic proteins cleaved-PARP and BAX and downregulation of antiapoptotic proteins Bcl-2 and p-AKT by western blot, especially in the presence of the autophagy inhibitor CQ. The results showed that the apoptosis of gastric cancer cells caused by C21 fraction was enhanced by inhibiting autophagy. The current findings reveal a new mechanism for the antitumor activity of C21 fraction on gastric cancer.
Collapse
Affiliation(s)
- Kaiqiang Li
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Ke Hao
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Yu Zhang
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Aibo Xu
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Qianni Wang
- School
of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yaoqiang Du
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Lingling Wu
- School
of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Bingyu Chen
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Wei Zhang
- Research
Center of Blood Transfusion Medicine, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Zhen Wang
- School
of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
6
|
Gebril SM, Ito Y, Abu-Dief EE, Hussein MRA, Elsayed HM, Mohammad AN, Abdelaal UM, Higuchi K. Ultra-structural study of the indomethacin-induced apoptosis and autophagy in rat gastric parietal cells. Ultrastruct Pathol 2020; 44:300-313. [PMID: 32672114 DOI: 10.1080/01913123.2020.1772429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND AIM OF THE WORK Indomethacin (IND), a non-steroidal anti-inflammatory drug, can induce gastric mucosal ulcerations. To date, the ultra-structural changes in the parietal cells (PCs) of the gastric mucosa following the intake of IND are mostly unknown. We carried out the current investigation to get insights into this issue. MATERIALS AND METHODS We established an animal model consisting of 35 adult male Sprague Dawley rats. The animals were divided into three groups, including; control (normal feeding), fasting, and indomethacin-treated groups. After treatment of 18-h fasting rats with IND, they were sacrificed at 3, 6, and 12-h intervals. The morphological features, including the apoptotic, and autophagic changes in the gastric mucosa PCs were examined using transmission electron microscopy. RESULTS In normal feeding animals (control group), the gastric PCs were present in various stages of activity. Fasting was associated with the predominance of the inactive parietal cells with features of up-regulated autophagy. In the IND -treated animals (at 3-h interval), PCs showed prominent autophagic changes, and subtle apoptotic cell death. In the IND -treated animals (at 6-12-h interval), PCs showed prominent apoptotic changes, and subtle autophagic features. CONCLUSIONS Our study indicates that IND treatment could induce gastropathy through time-dependent alterations in the autophagic and apoptotic machinery of PCs. Further studies are needed to examine the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sahar M Gebril
- Department of Anatomy, and Cell Biology, Osaka Medical College , Takatsuki, Japan.,Department of Histology, Faculty of Medicine, Sohag University , Sohag, Egypt
| | - Yuko Ito
- Department of Anatomy, and Cell Biology, Osaka Medical College , Takatsuki, Japan
| | - Eman E Abu-Dief
- Department of Histology, Faculty of Medicine, Sohag University , Sohag, Egypt
| | | | - Hoda M Elsayed
- Department of Histology, Faculty of Medicine, Sohag University , Sohag, Egypt
| | - Asmaa Naser Mohammad
- Department of Tropical Medicine and Gastroenterology, Sohag University Hospital , Sohag, Egypt
| | - Usama M Abdelaal
- Department of Internal Medicine, Sohag University Hospital , Egypt.,Department of Internal Medicine, Osaka Medical College , Takatsuki, Japan
| | - Kazuhide Higuchi
- Department of Internal Medicine, Osaka Medical College , Takatsuki, Japan
| |
Collapse
|
7
|
Wang Y, Sun L, Qiu W, Qi W, Qi Y, Liu Z, Liu S, Lv J. Inhibiting Forkhead box K1 induces autophagy to reverse epithelial-mesenchymal transition and metastasis in gastric cancer by regulating Myc-associated zinc finger protein in an acidic microenvironment. Aging (Albany NY) 2020; 12:6129-6150. [PMID: 32268297 PMCID: PMC7185099 DOI: 10.18632/aging.103013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
Background: Forkhead box K1 (FOXK1) is a transcription factor belonging to the Forkhead box (FOX) family and is closely related to the development of various cancers, but the functional mechanism through which FOXK1 regulates autophagy and epithelial-mesenchymal transition (EMT) in the acidic microenvironment of gastric cancer (GC) remains unclear. Results: Our results indicated that the inhibition of FOXK1 induced autophagy and thus exerted antimetastatic effects in an acidic microenvironment. The dual inhibition of mammalian target of rapamycin (mTOR) and FOXK1 enhanced autophagy and reversed EMT of acidic GC cells. In addition, FOXK1 activated transcription in conjunction with the MAZ promoter. Conclusion: Together, our results suggest that FOXK1 can be used as an independent prognostic indicator for GC patients. We also revealed a new strategy involving the cotargeting of FOXK1 and autophagy to reverse the effects of EMT. MAZ is involved in the development and progression of GC as a downstream target of FOXK1. Methods: Here, the cellular responses to the inhibition of FOXK1 in GC were studied in vivo and in vitro through wound healing assays, transwell assays, Western blotting, laser confocal microscopy and transmission electron microscopy. The molecular mechanisms of FOXK1 and Myc-associated zinc finger protein (MAZ) were studied via chromatin immunoprecipitation sequencing (ChIP-seq), bioinformatics, Western blotting, and quantitative real-time PCR (q-PCR).
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Libin Sun
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Wensheng Qiu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Weiwei Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Yaoyue Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Zhao Liu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Shihai Liu
- Central Laboratory, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| | - Jing Lv
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266071, Shandong, China
| |
Collapse
|
8
|
Po WW, Thein W, Khin PP, Khing TM, Han KWW, Park CH, Sohn UD. Fluoxetine Simultaneously Induces Both Apoptosis and Autophagy in Human Gastric Adenocarcinoma Cells. Biomol Ther (Seoul) 2020; 28:202-210. [PMID: 31522488 PMCID: PMC7059812 DOI: 10.4062/biomolther.2019.103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 01/14/2023] Open
Abstract
Fluoxetine is used widely as an antidepressant for the treatment of cancer-related depression, but has been reported to also have anti-cancer activity. In this study, we investigated the cytotoxicity of fluoxetine to human gastric adenocarcinoma cells; as shown by the MTT assay, fluoxetine induced cell death. Subsequently, cells were treated with 10 or 20 µM fluoxetine for 24 h and analyzed. Apoptosis was confirmed by the increased number of early apoptotic cells, shown by Annexin V- propidium iodide staining. Nuclear condensation was visualized by DAPI staining. A significant increase in the expression of cleaved PARP was observed by western blotting. The pan-caspase inhibitor Z-VAD-FMK was used to detect the extent of caspase-dependent cell death. The induction of autophagy was determined by the formation of acidic vesicular organelles (AVOs), which was visualized by acridine orange staining, and the increased expression of autophagy markers, such as LC3B, Beclin 1, and p62/SQSTM 1, observed by western blotting. The expression of upstream proteins, such as p-Akt and p-mTOR, were decreased. Autophagic degradation was evaluated by using bafilomycin, an inhibitor of late-stage autophagy. Bafilomycin did not significantly enhance LC3B expression induced by fluoxetine, which suggested autophagic degradation was impaired. In addition, the co-administration of the autophagy inhibitor 3-methyladenine and fluoxetine significantly increased fluoxetine-induced apoptosis, with decreased p-Akt and markedly increased death receptor 4 and 5 expression. Our results suggested that fluoxetine simultaneously induced both protective autophagy and apoptosis and that the inhibition of autophagy enhanced fluoxetine-induced apoptosis through increased death receptor expression.
Collapse
Affiliation(s)
- Wah Wah Po
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Wynn Thein
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Phyu Phyu Khin
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tin Myo Khing
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Khin Wah Wah Han
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Chan Hee Park
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.,Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Uy Dong Sohn
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
9
|
Heo G, Kang D, Park C, Kim SJ, Choo J, Lee Y, Yoo JW, Jung Y, Lee J, Kim ND, Chung HY, Moon HR, Im E. Pro-apoptotic effect of the novel benzylidene derivative MHY695 in human colon cancer cells. Oncol Lett 2019; 18:3256-3264. [PMID: 31452803 PMCID: PMC6704326 DOI: 10.3892/ol.2019.10664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 06/20/2019] [Indexed: 12/29/2022] Open
Abstract
The induction of apoptosis is a useful strategy in anti-cancer research. Various Moon Hyung Yang (MHY) compounds have been developed as novel anti-cancer drug candidates; in the present study, the pro-apoptotic effects of (Z)-5-(3-ethoxy-4- hydroxybenzylidene)-2-thioxothiazolidin-4-one (MHY695) on HCT116 human colon cancer cells were assessed. MTT assays were performed to investigate the dose-dependent cytotoxic effects of MHY695 on HCT116 cells. Immunofluorescence staining and flow cytometry analyses were performed to identify apoptotic cell death, and western blot analysis was used to investigate the apoptotic-signaling pathways. A mouse xenograft model was also used to determine the effects of MHY695 in vivo. MHY695 decreased the viability of HCT116 cells and induced apoptotic cytotoxicity. The apoptotic mechanisms induced by MHY695 involved the dephosphorylation of Bcl-2-associated agonist of cell death protein following protein kinase B inactivation, induced myeloid leukaemia cell differentiation protein and BH3-interacting domain death agonist truncation, caspase-3 and -9 activation and poly (ADP-ribose) polymerase cleavage. In addition, MHY695 significantly suppressed tumor growth in the mouse xenograft model, compared with the vehicle control. Notably, MHY695 exhibited potent anti-cancer effects in four different types of human colon cancer cell line, including Caco-2, DLD-1, HT-29 and HCT116. Additionally, MHY695 showed reduced cytotoxicity in NCM460, normal colonic epithelial cells. Furthermore, MHY-induced cytotoxicity in colon cancer cells was independent of the tumor suppressor protein p53. Collectively, these observations suggested that MHY695 may be a novel drug for the treatment of colon cancer.
Collapse
Affiliation(s)
- Gwangbeom Heo
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Dongwan Kang
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Chaeun Park
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Su Jin Kim
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jieun Choo
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Yunna Lee
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jaewon Lee
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Nam Deuk Kim
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Eunok Im
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| |
Collapse
|
10
|
Kim MY, Kruger AJ, Jeong JY, Kim J, Shin PK, Kim SY, Cho JY, Hahm KB, Hong SP. Combination Therapy with a PI3K/mTOR Dual Inhibitor and Chloroquine Enhances Synergistic Apoptotic Cell Death in Epstein-Barr Virus-Infected Gastric Cancer Cells. Mol Cells 2019; 42:448-459. [PMID: 31085812 PMCID: PMC6602147 DOI: 10.14348/molcells.2019.2395] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
The phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway is a promising target for gastric cancer (GC) treatment; however the efficacy of PI3K/mTOR dual inhibitors in GC has not yet been maximized. Additionally, the effect of autophagy regulation by PI3K/mTOR dual inhibitors has not been clearly elucidated in GC treatment. We aimed to show that our newly developed PI3K/mTOR dual inhibitor, CMG002, when combined with an autophagy inhibitor, chloroquine (CQ), potently induces effective cancer cell death in Epstein–Barr virus (EBV)-associated gastric cancer (EBVaGC) cells, where both the PI3K/AKT/mTOR and autophagy pathways play important roles in disease pathogenesis. EBV- and mock-infected AGS and NUGC3 GC cell lines were treated with CMG002 +/− CQ. PI3K/AKT/mTOR signaling pathway mediators, cellular apoptosis and autophagy markers were confirmed by Western blot assay. Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay. CMG002 effectively blocked the PI3K/AKT/mTOR pathway by markedly decreasing phosphorylation of AKT and its downstream mediator S6. CMG002 induced G0/G1 cell cycle arrest and enhanced apoptotic cell death in AGS and NUGC3 cells, particularly EBV-infected cells compared with mock-infected cells, as confirmed by flow cytometric analyses and TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assays. The combination of CMG002 plus CQ synergistically increased apoptotic cell death in EBV-infected GC cell lines when compared with CMG002 alone (P < 0.05). Our results suggest that the new PI3K/mTOR dual inhibitor, CMG002, when used in combination with the autophagy inhibitor, CQ, provides enhanced therapeutic efficacy against EBVaGC.
Collapse
Affiliation(s)
- Mi-Young Kim
- Digestive Disease Center, CHA University, Seongnam 13496,
Korea
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,
USA
| | - Annie J. Kruger
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,
USA
- Division of Gastroenterology, MedStar Georgetown University Hospital, Washington, DC 20007,
USA
| | - Ju-Yeon Jeong
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam 13496,
Korea
| | - Jaehee Kim
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam 13496,
Korea
| | - Phil kyung Shin
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam 13496,
Korea
| | - Sun Young Kim
- Department of Hematology and Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351,
Korea
| | - Joo Young Cho
- Digestive Disease Center, CHA University, Seongnam 13496,
Korea
| | - Ki Baik Hahm
- Digestive Disease Center, CHA University, Seongnam 13496,
Korea
| | - Sung Pyo Hong
- Digestive Disease Center, CHA University, Seongnam 13496,
Korea
| |
Collapse
|
11
|
The cytotoxicity of some phenanthroline-based antimicrobial copper(II) and ruthenium(II) complexes. J Inorg Biochem 2018; 180:61-68. [DOI: 10.1016/j.jinorgbio.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 02/02/2023]
|
12
|
Chang WT, Liu W, Chiu YH, Chen BH, Chuang SC, Chen YC, Hsu YT, Lu MJ, Chiou SJ, Chou CK, Chiu CC. A 4-Phenoxyphenol Derivative Exerts Inhibitory Effects on Human Hepatocellular Carcinoma Cells through Regulating Autophagy and Apoptosis Accompanied by Downregulating α-Tubulin Expression. Molecules 2017; 22:molecules22050854. [PMID: 28531143 PMCID: PMC6154338 DOI: 10.3390/molecules22050854] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cancer worldwide. Advanced HCCs are usually resistant to anticancer drugs, causing unsatisfactory chemotherapy outcomes. In this study, we showed that a 4-phenoxyphenol derivative, 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), exerts an inhibitory activity against two HCC cell lines, Huh7 and Ha22T. We further investigated the anti-HCC activities of 4-HPPP, including anti-proliferation and induction of apoptosis. Our results showed that higher dosage of 4-HPPP downregulates the expression of α-tubulin and causes nuclear enlargement in both the Huh-7 and Ha22T cell lines. Interestingly, the colony formation results showed a discrepancy in the inhibitory effect of 4-HPPP on HCC and rat liver epithelial Clone 9 cells, suggesting the selective cytotoxicity of 4-HPPP toward HCC cells. Furthermore, the cell proliferation and apoptosis assay results illustrated the differences between the two HCC cell lines. The results of cellular proliferation assays, including trypan blue exclusion and colony formation, revealed that 4-HPPP inhibits the growth of Huh7 cells, but exerts less cytotoxicity in Ha22T cells. Furthermore, the annexin V assay performed for detecting the apoptosis showed similar results. Western blotting results showed 4-HPPP caused the increase of pro-apoptotic factors including cleaved caspase-3, Bid and Bax in HCC cells, especially in Huh-7. Furthermore, an increase of autophagy-associated protein microtubule-associated protein-1 light chain-3B (LC3B)-II and the decrease of Beclin-1 and p62/SQSTM1 were observed following 4-HPPP treatment. Additionally, the level of γH2A histone family, member X (γH2AX), an endogenous DNA damage biomarker, was dramatically increased in Huh7 cells after 4-HPPP treatment, suggesting the involvement of DNA damage pathway in 4-HPPP-induced apoptosis. On the contrary, the western blotting results showed that treatment up-regulates pro-survival proteins, including the phosphorylation of protein kinase B (Akt) and the level of survivin on Ha22T cells, which may confer a resistance toward 4-HPPP. Notably, the blockade of extracellular signal-regulated kinases (ERK), but not Akt, enhanced the cytotoxicity of 4-HPPP against Ha22T cells, indicating the pro-survival role of ERK in 4-HPPP-induced anti-HCC effect. Our present work suggests that selective anti-HCC activity of 4-HPPP acts through induction of DNA damage. Accordingly, the combination of ERK inhibitor may significantly enhance the anti-cancer effect of 4-HPPP for those HCC cells which overexpress ERK in the future.
Collapse
Affiliation(s)
- Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yi-Han Chiu
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yi-Lan 266, Taiwan.
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Transplantation Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yun-Tzh Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Mei-Jei Lu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shean-Jaw Chiou
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chon-Kit Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Translational Research Center, Cancer Center, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
13
|
Qian HR, Yang Y. Functional role of autophagy in gastric cancer. Oncotarget 2017; 7:17641-51. [PMID: 26910278 PMCID: PMC4951239 DOI: 10.18632/oncotarget.7508] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/06/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a highly regulated catabolic pathway responsible for the degradation of long-lived proteins and damaged intracellular organelles. Perturbations in autophagy are found in gastric cancer. In host gastric cells, autophagy can be induced by Helicobacter pylori (or H. pylori) infection, which is associated with the oncogenesis of gastric cancer. In gastric cancer cells, autophagy has both pro-survival and pro-death functions in determining cell fate. Besides, autophagy modulates gastric cancer metastasis by affecting a wide range of pathological events, including extracellular matrix (ECM) degradation, epithelial-to-mesenchymal transition (EMT), tumor angiogenesis, and tumor microenvironment. In addition, some of the autophagy-related proteins, such as Beclin 1, microtubule-associated protein 1 light chain 3 (MAP1-LC3), and p62/sequestosome 1 (SQSTM1) have certain prognostic values for gastric cancer. In this article, we review the recent studies regarding the functional role of autophagy in gastric cancer.
Collapse
Affiliation(s)
- Hao-ran Qian
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yi Yang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| |
Collapse
|