1
|
Zhong Y, Shuai Y, Yang J, Zhang M, He T, Zheng L, Yang S, Peng S. LOC730101 improves ovarian cancer drug sensitivity by inhibiting autophagy-mediated DNA damage repair via BECN1. Cell Death Dis 2024; 15:893. [PMID: 39695078 DOI: 10.1038/s41419-024-07278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Drug resistance and recurrence are still the bottlenecks in the clinical treatment of ovarian cancer (OC), seriously affecting patients' prognosis. Therefore, it is an urgent challenge for OC to be overcome towards precision therapy by studying the mechanism of OC drug resistance, finding new drug resistance targets and developing new effective treatment strategies. In this study, we found that lncRNA LOC730101 played an essential role in attenuating drug resistance in OC. LOC730101 was significantly down-regulated in platinum-resistant ovarian cancer tissues, and ectopic overexpression of LOC730101 substantially increased chemotherapy-induced apoptosis. Mechanistically, LOC730101 specifically binds to BECN1 and inhibits the formation of autophagosome BECN1/VPS34 by reducing phosphorylation of BECN1, thereby inhibiting autophagy and promoting drug sensitivity in ovarian cancer cells following treatment with cisplatin and PARP inhibitors. Moreover, LOC730101 inhibits the expression and activity of RNF168 via p62, which in turn affects H2A ubiquitination-mediated DNA damage repair and promotes drug sensitivity in ovarian cancer cells. Our findings demonstrated that LOC730101 played an important role in regulating the formation of the autophagic complex and that inhibition of autophagy significantly enhances the drug sensitivity of OC. And LOC730101 may be used as a prognostic marker to predict the sensitivity of OC to platinum and PARP inhibitors.
Collapse
Affiliation(s)
- Yancheng Zhong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Key laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, China
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yang Shuai
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Yang
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mojian Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Tiantian He
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Leliang Zheng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Sheng Yang
- The Reproductive Medicine Center, The Third Affiliated Hospital of ShenZhen University, Shenzhen Luohu Hospital Group, Shenzhen, China.
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital and School of Basic Medical Science, Central South University, Changsha, Hunan, China.
- Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Insight into autophagy in platinum resistance of cancer. Int J Clin Oncol 2023; 28:354-362. [PMID: 36705869 DOI: 10.1007/s10147-023-02301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023]
Abstract
Platinum drugs, as a class of widely used chemotherapy agents, frequently appear in the treatment of cancer at different phrases. However, platinum resistance is the major bottleneck of platinum drugs for exerting anti-tumor effect. At present, the mechanism of platinum resistance has been thoroughly explored in terms of drug delivery methods, DNA damage repair function, etc., but it has not yet been translated into an effective weapon for reversing platinum resistance. Recently, autophagy has been proved to be closely related to platinum resistance, and the involved molecular mechanism may provide a new perspective on platinum resistance. The aim of this review is to sort out the studies related to autophagy and platinum resistance, and to focus on summarizing the relevant molecular mechanisms, so as to provide clues for future studies related to autophagy and platinum resistance.
Collapse
|
3
|
Tam C, Rao S, Waye MMY, Ng TB, Wang CC. Autophagy signals orchestrate chemoresistance of gynecological cancers. Biochim Biophys Acta Rev Cancer 2021; 1875:188525. [PMID: 33600824 DOI: 10.1016/j.bbcan.2021.188525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Gynecological cancers are characterized by a high mortality rate when chemoresistance develops. Autophagy collaborates with apoptosis and participates in homeostasis of chemoresistance. Recent findings supported that crosstalk of necrotic, apoptotic and autophagic factors, and chemotherapy-driven hypoxia, oxidative stress and ER stress play critical roles in chemoresistance in gynecological cancers. Meanwhile, current studies have shown that autophagy could be regulated by and cooperate with metabolic regulator, survival factors, stemness factors and specific post-translation modification in chemoresistant tumor cells. Meanwhile, non-coding RNA and autophagy crosstalk also contribute to the chemoresistance. Until now, analysis of individual autophagy factors towards the clinical significance and chemoresistance in gynecological cancer is still lacking. We suggest comprehensive integrated analysis of cellular homeostasis and tumor microenvironment to clarify the role of autophagy and the associated factors in cancer progression and chemoresistance. Panel screening of pan-autophagic factors will pioneer the development of risk models for predicting efficacy of chemotherapy and guidelines for systematic treatment and precision medicine.
Collapse
Affiliation(s)
- Chit Tam
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.
| | - Shitao Rao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; School of Medical Technology and Engineering, Fujian Medical University, Fujian, China
| | - Mary Miu Yee Waye
- The Nethersole School of Nursing, The Chinese University of Hong Kong, Hong Kong, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; Reproduction and Development Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Liu T, Zhang J, Li K, Deng L, Wang H. Combination of an Autophagy Inducer and an Autophagy Inhibitor: A Smarter Strategy Emerging in Cancer Therapy. Front Pharmacol 2020; 11:408. [PMID: 32322202 PMCID: PMC7156970 DOI: 10.3389/fphar.2020.00408] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
Autophagy is considered a cytoprotective function in cancer therapy under certain conditions and is a drug resistance mechanism that represents a clinical obstacle to successful cancer treatment and leads to poor prognosis in cancer patients. Because certain clinical drugs and agents in development have cytoprotective autophagy effects, targeting autophagic pathways has emerged as a potential smarter strategy for cancer therapy. Multiple preclinical and clinical studies have demonstrated that autophagy inhibition augments the efficacy of anticancer agents in various cancers. Autophagy inhibitors, such as chloroquine and hydroxychloroquine, have already been clinically approved, promoting drug combination treatment by targeting autophagic pathways as a means of discovering and developing more novel and more effective cancer therapeutic approaches. We summarize current studies that focus on the antitumor efficiency of agents that induce cytoprotective autophagy combined with autophagy inhibitors. Furthermore, we discuss the challenge and development of targeting cytoprotective autophagy as a cancer therapeutic approach in clinical application. Thus, we need to facilitate the exploitation of appropriate autophagy inhibitors and coadministration delivery system to cooperate with anticancer drugs. This review aims to note optimal combination strategies by modulating autophagy for therapeutic advantage to overcome drug resistance and enhance the effect of antitumor therapies on cancer patients.
Collapse
Affiliation(s)
- Ting Liu
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhang
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kangdi Li
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Lingnan Deng
- Department of Digestion, The Second Affiliated Hospital of Jiangxi University TCM, Nanchang, China
| | - Hongxiang Wang
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Sagrillo-Fagundes L, Bienvenue-Pariseault J, Vaillancourt C. Melatonin: The smart molecule that differentially modulates autophagy in tumor and normal placental cells. PLoS One 2019; 14:e0202458. [PMID: 30629581 PMCID: PMC6328125 DOI: 10.1371/journal.pone.0202458] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Melatonin has protective roles in normal cells and cytotoxic actions in cancer cells, with effects involving autophagy and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor pathways. Hypoxia/reoxygenation (H/R) induces oxidative damage and apoptosis. These consequences activate autophagy, which degrades damaged cellular content, as well as activates Nrf2 the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor, and thereby the expression of protective genes. Melatonin has protective roles in normal cells and cytotoxic actions in cancer cells, with effects involving autophagy and Nrf2 pathways. The current study shows melatonin to differentially modulate autophagy and Nrf2 pathways in tumor and normal placental cells exposed to H/R. BeWo, a human placental choriocarcinoma cell line, and primary villous cytotrophoblasts isolated from normal term placenta, were maintained in normoxia (8% O2) for 24 h or exposed to hypoxia (0.5% of O2 for 4 h) followed by 20 h of normoxia, creating a situation of H/R, in the presence or absence of 1 mM melatonin. Melatonin induced a 7-fold increase in the activation of 5' adenosine monophosphate-activated protein kinase (AMPK)α, an upstream modulator of autophagy, rising to a 16-fold increase in BeWo cells co-exposed to H/R and melatonin, compared to controls. H/R induced autophagosome formation via the increased expression of Beclin-1 (by 94%) and ATG7 (by 97%) in BeWo cells. Moreover, H/R also induced autophagic activity, indicated by the by the 630% increase in P62, and increased Nrf2 by 314% in BeWo cells. In H/R conditions, melatonin reduced autophagic activity by 74% and Nrf2 expression activation by 300%, leading to BeWo cell apoptosis. In contrast, In human primary villous cytotrophoblasts, H/R induced autophagy and Nrf2, which melatonin further potentiated, thereby affording protection against H/R. This study demonstrates that melatonin differentially modulates autophagy and the Nrf2 pathway in normal vs. tumor trophoblast cells, being cytoprotective in normal cells whilst increasing apoptosis in tumoral trophoblast cells.
Collapse
Affiliation(s)
- Lucas Sagrillo-Fagundes
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
| | - Josianne Bienvenue-Pariseault
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
| | - Cathy Vaillancourt
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
- * E-mail:
| |
Collapse
|
6
|
Li W, Zhang H, Yang L, Wang Y. Cancerous inhibitor of protein phosphatase 2A regulates cisplatin resistance in ovarian cancer. Oncol Lett 2018; 17:1211-1216. [PMID: 30655886 DOI: 10.3892/ol.2018.9653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/06/2018] [Indexed: 11/05/2022] Open
Abstract
Ovarian cancer is the most aggressive type of gynecological cancer. The cause of the poor survival rate is the development of chemotherapy resistance to platinum-based therapies, including cisplatin. The present study aimed to investigate the mechanism of cancerous inhibitor of protein phosphatase 2A (CIP2A)-induced chemoresistance in ovarian cancer. The present study initially investigated the expression of CIP2A in the ovarian tumor tissue, cisplatin-sensitive SKOV-3 cell line, and cisplatin-resistant ovarian carcinoma SKOV-3CDDP/R cell line. In addition, CIP2A was knocked down using small interference RNA in ovarian cancer cells and the chemosensitivity of these cells was analyzed. The results demonstrated that CIP2A expression was significantly higher in patients with ovarian cancer and in the cisplatin-resistant ovarian carcinoma SKOV-3CDDP/R cell line at the mRNA and protein levels. The proliferation and chemosensitivity were decreased and enhanced, respectively, when CIP2A was knocked down. CIP2A silencing significantly promoted the apoptosis induced by cisplatin in SKOV-3CDDP/R cells, suggesting that CIP2A participated in the cisplatin resistance of ovarian cancer cells and that CIP2A silencing enhanced the apoptosis induced by cisplatin. CIP2A may be considered as a potential candidate for modulating cisplatin therapy in ovarian cancer.
Collapse
Affiliation(s)
- Wanbin Li
- Department of Gynecology, Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Hongyan Zhang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Linqing Yang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Yunfei Wang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| |
Collapse
|
7
|
Knockdown of Yin Yang 1 enhances anticancer effects of cisplatin through protein phosphatase 2A-mediated T308 dephosphorylation of AKT. Cell Death Dis 2018; 9:747. [PMID: 29970878 PMCID: PMC6030060 DOI: 10.1038/s41419-018-0774-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/23/2018] [Accepted: 05/28/2018] [Indexed: 11/16/2022]
Abstract
Cisplatin is still one of the first-line drugs for chemotherapy of head and neck squamous cell carcinoma (HNSCC) and shows a survival advantage for HNSCC. However, a substantial proportion of HNSCC eventually becomes resistance to cisplatin and the underlying mechanisms remain to be fully understood. Yin Yang 1 (YY1) is a multifunctional protein regulating both gene transcription and protein modifications and also plays a role in chemotherapy resistance. Here, we reported that knockdown of YY1 by lentivirus-mediated short hairpin RNA or tetracycline-inducible short hairpin RNA enhanced cisplatin-induced apoptosis and inhibition of cell proliferation, migration and invasion in the HNSCC cell lines, and inhibition of the xenograft tumor growth. The underlying mechanisms were revealed that knockdown of YY1 downregulated both S473 and T308 phosphorylation of AKT (protein kinase B), which was mainly responsible for cisplatin resistance, whereas overexpression of YY1 upregulated both S473 and T308 phosphorylation. Cisplatin upregulated YY1 mRNA and protein expression and both S473 and T308 phosphorylation of AKT. In the presence of cisplatin, knockdown of YY1 not only blocked cisplatin-induced increase in S473 and T308 phosphorylation of AKT, but still downregulated T308 phosphorylation. Moreover, protein phosphatase 2A (PP2A) antagonist, okadaic acid, upregulated T308, but not S473, phosphorylation, and simultaneously abolished YY1 knockdown-mediated enhancement of cisplatin-induced inhibition of cell proliferation. In addition, knockdown of YY1 promoted PP2A activity through upregulating mRNA and protein expressions of PP2A catalytic subunit alpha (PPP2CA) through the binding of YY1 in the promoter of PPP2CA. Conversely, activating PP2A by forskolin also promoted YY1 degradation and subsequently inhibited T308 phosphorylation. These results suggested that knockdown of YY1 enhanced anticancer effects of cisplatin through PP2A mediating T308 dephosphorylation of AKT, and that targeting YY1 or PP2A would enhance the efficiency of cisplatin chemotherapy in treatment of HNSCC.
Collapse
|
8
|
Zhan L, Zhang Y, Wang W, Song E, Fan Y, Li J, Wei B. Autophagy as an emerging therapy target for ovarian carcinoma. Oncotarget 2018; 7:83476-83487. [PMID: 27825125 PMCID: PMC5347782 DOI: 10.18632/oncotarget.13080] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/21/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a conserved cellular self-digestion pathway for maintenance of homeostasis under basal and stressed conditions. Autophagy plays pivotal roles in the pathogenesis of many diseases, such as aging-related diseases, autoimmune diseases, cardiovascular diseases, and cancers. Of special note is that accumulating data suggest an intimate relationship between autophagy and ovarian carcinoma. Autophagy is well identified to act as either as a tumor-suppressor or as a tumor-promoter in ovarian carcinoma. The exact function of autophagy in ovarian carcinoma is highly dependent on the circumstances of cancer including hypoxic, nutrient-deficient, chemotherapy and so on. However, the mechanism underlying autophagy associated with ovarian carcinoma remains elusive, the precise role of autophagy in ovarian carcinoma also remains undetermined. In this review, we tried to sum up and discuss recent research achievements of autophagy in ovarian cancer. Moreover, waves of novel therapies ways for ovarian carcinoma based on the functions of autophagy were collected.
Collapse
Affiliation(s)
- Lei Zhan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu Zhang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenyan Wang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Enxue Song
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yijun Fan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China
| | - Bing Wei
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
9
|
Niu Q, Zhao W, Wang J, Li C, Yan T, Lv W, Wang G, Duan W, Zhang T, Wang K, Zhou D. LicA induces autophagy through ULK1/Atg13 and ROS pathway in human hepatocellular carcinoma cells. Int J Mol Med 2018; 41:2601-2608. [PMID: 29484365 PMCID: PMC5846666 DOI: 10.3892/ijmm.2018.3499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/07/2018] [Indexed: 11/05/2022] Open
Abstract
Chemotherapy is the best choice for the vast majority of hepatocellular carcinoma patients at late stage, but few effective chemotherapy drugs are available in clinic. Licochalcone A (LicA) is a new chemotherapy drug inducing apoptosis as Bcl-2 inhibitor, but few studies report on LicA-induced autophagy. This study investigated the phenomenon and mechanisms of LicA-induced autophagy looking for a targeted combination drug. Human hepatocellular carcinoma cells (HCCs) were treated with LicA, to detect markers of autophagy and to investigate the mechanisms. In order to investigate the role of reactive oxygen species (ROS) in LicA-induced autophagy, ROS, glutathione (GSH) and O2− were measured in LicA treated HCCs, and antioxidant N-Acetyl-L-cysteine (NAC) was cotreated with LicA in HCCs, then mechanisms of ROS-induced autophagy was investigated in LicA or LicA combined with NAC treated HCCs. Finally, the LicA-induced apoptosis was detected in LicA combined with NAC treated HCCs. We first report that LicA can induce autophagy through ULK1/Atg13 and ROS pathway in HCCs, suppression of LicA-induced ROS through antioxidant NAC can enhance LicA-induced apoptosis, promoting the function of LicA killing HCCs. LicA can activate the ULK1/Atg13 complex which is upstream of autophagy, additionally, LicA also can promote ROS generation, ROS trigger the expression level of TSC1/2 complex, PRAS40, CTMP, PP2A, PDK1 and Rubicon change, these molecules are upstream of autophagy. In conclusion, LicA can induce autophagy through ULK1/Atg13 and ROS pathway in HCCs, LicA combined with NAC can enhance LicA-induced apoptosis. Our results may provide a novel design for clinical hepatocellular carcinoma therapy trials.
Collapse
Affiliation(s)
- Qiang Niu
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Wei Zhao
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Jin Wang
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Chunming Li
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Tao Yan
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Wei Lv
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Guojing Wang
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Weihong Duan
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Tao Zhang
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Kunnan Wang
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| | - Dinghua Zhou
- Department of Hepatobiliary Surgery, Rocket Army General Hospital, Beijing 100088, P.R. China
| |
Collapse
|
10
|
Phosphatases and solid tumors: focus on glioblastoma initiation, progression and recurrences. Biochem J 2017; 474:2903-2924. [PMID: 28801478 DOI: 10.1042/bcj20170112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
Phosphatases and cancer have been related for many years now, as these enzymes regulate key cellular functions, including cell survival, migration, differentiation and proliferation. Dysfunctions or mutations affecting these enzymes have been demonstrated to be key factors for oncogenesis. The aim of this review is to shed light on the role of four different phosphatases (PTEN, PP2A, CDC25 and DUSP1) in five different solid tumors (breast cancer, lung cancer, pancreatic cancer, prostate cancer and ovarian cancer), in order to better understand the most frequent and aggressive primary cancer of the central nervous system, glioblastoma.
Collapse
|
11
|
FTY720 Induces Autophagy-Associated Apoptosis in Human Oral Squamous Carcinoma Cells, in Part, through a Reactive Oxygen Species/Mcl-1-Dependent Mechanism. Sci Rep 2017; 7:5600. [PMID: 28717222 PMCID: PMC5514089 DOI: 10.1038/s41598-017-06047-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 06/07/2017] [Indexed: 11/14/2022] Open
Abstract
In this study, we interrogated the mechanism by which the immunosuppressant FTY720 mediates anticancer effects in oral squamous cell carcinoma (OSCC) cells. FTY720 differentially suppressed the viability of the OSCC cell lines SCC4, SCC25, and SCC2095 with IC50 values of 6.1, 6.3, and 4.5 μM, respectively. This antiproliferative effect was attributable to the ability of FTY720 to induce caspase-dependent apoptosis. Mechanistic evidence suggests that FTY720-induced apoptosis was associated with its ability to inhibit Akt-NF-κB signaling, to facilitate the proteasomal degradation of the antiapoptotic protein Mcl-1, and to increase reactive oxygen species (ROS) generation. Both overexpression of Mcl-1 and inhibition of ROS partially protected cells from FTY720-induced caspase-9 activation, PARP cleavage and cytotoxicity. In addition, FTY720 induced autophagy in OSCC cells, as manifested by LC3B-II conversion, decreased p62 expression, and accumulation of autophagosomes. Inhibition of autophagy by bafilomycin A1 protected cells from FTY720-induced apoptosis. Together, these findings suggest an intricate interplay between autophagy and apoptosis in mediating the tumor-suppressive effect in OSCC cells, which underlies the translational potential of FTY720 in fostering new therapeutic strategies for OSCC.
Collapse
|
12
|
Zheng X, Chen W, Hou H, Li J, Li H, Sun X, Zhao L, Li X. Ginsenoside 20(S)-Rg3 induced autophagy to inhibit migration and invasion of ovarian cancer. Biomed Pharmacother 2017; 85:620-626. [DOI: 10.1016/j.biopha.2016.11.072] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022] Open
|
13
|
Xu S, Duan P, Li J, Senkowski T, Guo F, Chen H, Romero A, Cui Y, Liu J, Jiang SW. Zinc Finger and X-Linked Factor (ZFX) Binds to Human SET Transcript 2 Promoter and Transactivates SET Expression. Int J Mol Sci 2016; 17:ijms17101737. [PMID: 27775603 PMCID: PMC5085766 DOI: 10.3390/ijms17101737] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/24/2016] [Accepted: 10/08/2016] [Indexed: 02/05/2023] Open
Abstract
SET (SE Translocation) protein carries out multiple functions including those for protein phosphatase 2A (PP2A) inhibition, histone modification, DNA repair, and gene regulation. SET overexpression has been detected in brain neurons of patients suffering Alzheimer's disease, follicle theca cells of Polycystic Ovary Syndrome (PCOS) patients, and ovarian cancer cells, indicating that SET may play a pathological role for these disorders. SET transcript 2, produced by a specific promoter, represents a major transcript variant in different cell types. In this study, we characterized the transcriptional activation of human SET transcript 2 promoter in HeLa cells. Promoter deletion experiments and co-transfection assays indicated that ZFX, the Zinc finger and X-linked transcription factor, was able to transactivate the SET promoter. A proximal promoter region containing four ZFX-binding sites was found to be critical for the ZFX-mediated transactivation. Mutagenesis study indicated that the ZFX-binding site located the closest to the transcription start site accounted for most of the ZFX-mediated transactivity. Manipulation of ZFX levels by overexpression or siRNA knockdown confirmed the significance and specificity of the ZFX-mediated SET promoter activation. Chromatin immunoprecipitation results verified the binding of ZFX to its cognate sites in the SET promoter. These findings have led to identification of ZFX as an upstream factor regulating SET gene expression. More studies are required to define the in vivo significance of this mechanism, and specifically, its implication for several benign and malignant diseases related to SET dysregulation.
Collapse
Affiliation(s)
- Siliang Xu
- The State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China.
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Jinping Li
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Tristan Senkowski
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Fengbiao Guo
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515000, China.
| | - Haibin Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515000, China.
| | - Alberto Romero
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Yugui Cui
- The State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China.
| | - Jiayin Liu
- The State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China.
| | - Shi-Wen Jiang
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, USA.
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
14
|
Wesselborg S, Stork B. Autophagy signal transduction by ATG proteins: from hierarchies to networks. Cell Mol Life Sci 2015; 72:4721-57. [PMID: 26390974 PMCID: PMC4648967 DOI: 10.1007/s00018-015-2034-8] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/13/2015] [Accepted: 08/31/2015] [Indexed: 02/07/2023]
Abstract
Autophagy represents an intracellular degradation process which is involved in both cellular homeostasis and disease settings. In the last two decades, the molecular machinery governing this process has been characterized in detail. To date, several key factors regulating this intracellular degradation process have been identified. The so-called autophagy-related (ATG) genes and proteins are central to this process. However, several additional molecules contribute to the outcome of an autophagic response. Several review articles describing the molecular process of autophagy have been published in the recent past. In this review article we would like to add the most recent findings to this knowledge, and to give an overview of the network character of the autophagy signaling machinery.
Collapse
Affiliation(s)
- Sebastian Wesselborg
- Institute of Molecular Medicine I, Heinrich-Heine-University, Universitätsstr. 1, Building 23.12, 40225, Düsseldorf, Germany
| | - Björn Stork
- Institute of Molecular Medicine I, Heinrich-Heine-University, Universitätsstr. 1, Building 23.12, 40225, Düsseldorf, Germany.
| |
Collapse
|
15
|
Vizcaíno C, Rodríguez-Sánchez MA, Núñez LE, Morís F, Portugal J. Cytotoxic effects of mithramycin DIG-MSK can depend on the rise of autophagy. Toxicol In Vitro 2015; 29:1537-44. [PMID: 26079942 DOI: 10.1016/j.tiv.2015.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 11/29/2022]
Abstract
DIG-MSK (demycarosil-3D-β-D-digitoxosyl mithramycin SK; EC-8042), a novel analogue of mithramycin A, induced autophagy in HCT116 human colon carcinoma and, to a lesser extent, in A2780 human ovarian carcinoma cell lines, which was followed by apoptosis and/or necrotic cell death in a time-dependent way. The effects of DIG-MSK included changes in the expression of a set of genes involved in autophagy, the progression of cells through the different phases of cell cycle, and their halting at the checkpoints. Cells treated with the glucose analogue 2-DG (2-deoxy-D-glucose), which induces autophagy because it impairs cell metabolism, or co-treated with 2-DG plus DIG-MSK, also showed altered gene expression and autophagy. In A2780 cells, some genes involved in autophagy were down-regulated by the different treatments, yet the levels of the proteins they encode could be enough to ensure autophagic flux. In HCT116 cells, up-regulation of several pro-autophagic genes resulted in strong autophagic response. Acidic cell organelles and autophagic flux were more evident in HCT116 than in A2780 cells. DIG-MSK was still cytotoxic in cells that underwent autophagy induced by 2-DG. Therefore, we verified that autophagy resulting from a stress response did not protect cells against DIG-MSK, but, instead, autophagy promoted by either 2-DG or the novel mithralogue can enhance the antitumour activity, which depended on the cell type.
Collapse
Affiliation(s)
- Carolina Vizcaíno
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Barcelona, Spain
| | | | | | | | - José Portugal
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Barcelona, Spain.
| |
Collapse
|
16
|
Mao Y, Wang J, Zhao Y, Yan R, Li H, Chen CS, Lee RJ, Byrd JC, Lee LJ, Muthusamy N, Phelps MA. Quantification of OSU-2S, a novel derivative of FTY720, in mouse plasma by liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2014; 98:160-5. [PMID: 24927402 DOI: 10.1016/j.jpba.2014.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/01/2014] [Accepted: 05/06/2014] [Indexed: 02/03/2023]
Abstract
OSU-2S is a novel anti-cancer and immune modulatory agent designed specifically to avert the immunosuppressive effects and related toxicities observed in clinical studies with its predecessor analog, FTY720. To characterize its preclinical pharmacokinetics, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the quantification of OSU-2S in mouse plasma. Ethyl acetate extraction of samples containing OSU-2S and the internal standard, Sph-17, was followed by separation with a 6min gradient (water/0.1% formic acid and methanol/0.1% formic acid) on a reverse-phase C18 column at room temperature. Selected reaction monitoring was used for detection on a triple quadrupole mass spectrometer with positive ionization. The assay was linear over the concentration range 3-3000ng/mL with accuracy ranging from 103 to 111%, and both within- and between-run precision (CV%) ≤11%. All stability samples were within ±15% of nominal values, and replicates were within 15% CV. The assay was successfully applied to a mouse pharmacokinetic study of OSU-2S with intravenous and intraperitoneal administration. OSU-2S non-compartmental pharmacokinetic parameters, area under the concentration-time curve, clearance, and elimination half-life were estimated at 1522hμg/L, 3.06L/h/kg and 15.6h, respectively, for intravenous injection. Systemic availability after intraperitoneal injection was approximately 46%. These data demonstrate the OSU-2S compound displays acceptable pharmacokinetic properties for further in vivo pharmacologic evaluation, which can be facilitated by the validated LC-MS/MS assay.
Collapse
Affiliation(s)
- Yicheng Mao
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA
| | - Jiang Wang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yuan Zhao
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Ribai Yan
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Hao Li
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Ching-Shih Chen
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - L James Lee
- Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA; William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Mitch A Phelps
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Xing Y, Wang ZH, Ma DH, Han Y. FTY720 enhances chemosensitivity of colon cancer cells to doxorubicin and etoposide via the modulation of P-glycoprotein and multidrug resistance protein 1. J Dig Dis 2014; 15:246-59. [PMID: 24868599 DOI: 10.1111/1751-2980.12131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This study aimed to investigate the effects of FTY720 on inducing cell growth inhibition and enhancing the cytotoxicity of anti-cancer drugs in the human colon cancer cell line HCT-8 and its multidrug-resistant cell line HCT-8/5-fluorouracil (HCT-8/5-Fu). METHODS Cell viability and apoptosis after being treated with FTY720 alone or in combination with doxorubicin (DOX) and etoposide (VP16) were tested in HCT-8 and HCT-8/5-Fu cells. The changes in P-glycoprotein (P-gp) and multidrug resistance protein 1 (MRP1) were determined at the mRNA and functional levels. RESULTS FTY720 showed anti-proliferative activity against cancer cells in a dose-dependent and time-dependent manner and could enhance the cytotoxicity of DOX and VP16 in both HCT-8 and HCT-8/5-Fu cell lines. In addition, treatment with FTY720 resulted in the promotion of VP16-induced cell apoptosis and an increased accumulation of intracellular DOX and two specific fluorescent substrates of P-gp and MRP1 through the inhibition of efflux and the suppression of gene expression. CONCLUSION FTY720 exerts its chemosensitization effect in HCT-8 and HCT-8/5-Fu cell lines by promoting cell apoptosis and inhibiting P-gp and MRP1, which could be applied as a potential co-adjuvant therapeutic modality.
Collapse
|
18
|
FTY720 for cancer therapy (Review). Oncol Rep 2013; 30:2571-8. [PMID: 24100923 DOI: 10.3892/or.2013.2765] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/16/2013] [Indexed: 02/04/2023] Open
Abstract
2-Amino-2-[2-(4-octylphenyl)]-1,3-propanediol hydrochloride (FTY720) is a potent immunosuppressant which has been approved by the Food and Drug Administration (FDA) as a new treatment for multiple sclerosis. As an immunosuppressant, it displays its anti-multiple sclerosis, immunosuppressive effects by activating sphingosine-1-phosphate receptors (S1PRs). In addition to the immunosuppressive effects, FTY720 also shows preclinical antitumor efficacy in several cancer models. In most cases, phosphorylation of FTY720 is not required for its cytotoxic effect, indicating the involvement of S1PR-independent mechanisms which are starkly different from the immunosuppressive property of FTY720. In the present study, we reviewed the rapidly advancing field of FTY720 in cancer therapy as well as some molecular targets of the unphosphorylated form of FTY720.
Collapse
|