1
|
Rahdan F, Abedi F, Dianat-Moghadam H, Sani MZ, Taghizadeh M, Alizadeh E. Autophagy-based therapy for hepatocellular carcinoma: from standard treatments to combination therapy, oncolytic virotherapy, and targeted nanomedicines. Clin Exp Med 2024; 25:13. [PMID: 39621122 PMCID: PMC11611955 DOI: 10.1007/s10238-024-01527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Human hepatocellular carcinoma (HCC) has been identified as a significant cause of mortality worldwide. In recent years, extensive research has been conducted to understand the underlying mechanisms of autophagy in the pathogenesis of the disease, with the aim of developing novel therapeutic agents. Targeting autophagy with conventional therapies in invasive HCC has opened up new opportunities for treatment. However, the emergence of resistance and the immunosuppressive tumor environment highlight the need for combination therapy or specific targeting, as well as an efficient drug delivery system to ensure targeted tumor areas receive sufficient doses without affecting normal cells or tissues. In this review, we discuss the findings of several studies that have explored autophagy as a potential therapeutic approach in HCC. We also outline the potential and limitations of standard therapies for autophagy modulation in HCC treatment. Additionally, we discuss how different combination therapies, nano-targeted strategies, and oncolytic virotherapy could enhance autophagy-based HCC treatment in future research.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abedi
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
| | - Maryam Zamani Sani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Ma T, Li D, Yang ZR, Wang Q, Chen R, Lv N, Du K, Qin H, Tao J, Jiang H, Zhu J. Autophagy activator-loaded bicomponent peptide nanocarriers for phototherapy-triggered immunity enhancement against metastatic breast cancer. J Control Release 2024; 376:241-252. [PMID: 39384152 DOI: 10.1016/j.jconrel.2024.10.009] [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: 03/30/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/11/2024]
Abstract
Mild autophagy accompanied with immunogenic cell death (ICD) effect destructs immune-associated antigens, weakening the immune response against tumor growth. To address this dilemma, we develop a peptide-based bicomponent nanocarrier with encapsulation of a cellular hyperautophagy activator (STF-62247) for near-infrared (NIR) photo/immunotherapy to eliminate primary and metastatic breast tumors. The electrostatic-driven nanodrug (PPNPs@STF) with active-targeting and efficient endosomal escape can induce specific ICD effect upon NIR laser irradiation, and trigger autophagy to a mild activation state. Notably, the simultaneously released STF-62247 precisely promotes autophagy to an overactivated state, resulting in autophagic death of tumor cells and further boosting ICD-related antigen presentation. More importantly, the combined photo/immunotherapy of PPNPs@STF not only inhibits tumor cell proliferation, but also promotes dendritic cells (DCs)-associated immune response. In 4 T1 tumor-bearing mice, PPNPs@STF effectively inhibits growth of primary and distant tumors, and suppresses lung metastasis with a minimized side effect. This study provides a hyperautophagy activator-assisted strategy that can enhance ICD-based antitumor immune response for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Teng Ma
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Danqi Li
- Hubei Engineering Research Center for Skin Repair and Theranostics, Department of Dermatology, Union Hospital, Tongji Medical College, HUST, Wuhan 430022, China
| | - Zhuo-Ran Yang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Qi Wang
- Hubei Engineering Research Center for Skin Repair and Theranostics, Department of Dermatology, Union Hospital, Tongji Medical College, HUST, Wuhan 430022, China
| | - Rong Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Niannian Lv
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Kehan Du
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Huimin Qin
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Juan Tao
- Hubei Engineering Research Center for Skin Repair and Theranostics, Department of Dermatology, Union Hospital, Tongji Medical College, HUST, Wuhan 430022, China.
| | - Hao Jiang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| |
Collapse
|
3
|
Wang J, Liu M, Wang J, Li Z, Feng Z, Xu M, Wang H, Li H, Li Z, Yu J, Liu J, Wei Q, Zhang S, Zhang X. Zinc oxide nanoparticles with catalase-like nanozyme activity and near-infrared light response: A combination of effective photodynamic therapy, autophagy, ferroptosis, and antitumor immunity. Acta Pharm Sin B 2024; 14:4493-4508. [PMID: 39525585 PMCID: PMC11544279 DOI: 10.1016/j.apsb.2024.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
We prepared biocompatible and environment-friendly zinc oxide nanoparticles (ZnO NPs) with upconversion properties and catalase-like nanozyme activity. Photodynamic therapy (PDT) application is severely limited by the poor penetration of UV-Visible light and a hypoxic tumor environment. Here, we used ZnO NPs as a carrier for the photosensitizer chlorin e6 (Ce6) to construct zinc oxide-chlorin e6 nanoparticles (ZnO-Ce6 NPs), simultaneously addressing both problems. In terms of penetration, ZnO NPs convert 808 nm near-infrared light into 401 nm visible light to excite Ce6, achieving deep-penetrating photodynamic therapy under long-wavelength light. Interestingly, the ability to emit short-wavelength light under long-wavelength light is usually observed in upconversion nanoparticles. As nanozymes, ZnO NPs can catalyze the decomposition of hydrogen peroxide in tumors, providing oxygen for photodynamic action and relieving hypoxia. The enhanced photodynamic action produces a large amount of reactive oxygen species, which overactivate autophagy and trigger immunogenic cell death (ICD), leading to antitumor immunotherapy. In addition, even in the absence of light, ZnO and ZnO-Ce6 NPs can induce ferroptosis of tumor cells and exert antitumor effects.
Collapse
Affiliation(s)
- Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Man Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenhan Feng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhantao Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qingchao Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shuang Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
4
|
Ren Q, Dong Y, Huang Y, Xiao J, Ma Y, Liu Y, Sun H, Dai Y, Shi C, Wang S. Nrf2 induces angiogenesis in spinal cystic echinococcosis by activating autophagy via regulating oxidative stress. Biochem Pharmacol 2024; 226:116337. [PMID: 38844265 DOI: 10.1016/j.bcp.2024.116337] [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: 01/31/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Spinal cystic echinococcosis (CE) is a rare but malignant zoonosis that can cause disability or even death in more than half of patients. Due to the complex pathological features, it is not curable by conventional drugs and surgery, so new therapeutic targets urgently need to be discovered. In this study, we clarify the occurrence of the phenomenon of spinal encapsulation angiogenesis and explore its underlying molecular mechanisms. A co-culture system was established by protoscoleces (PSCs) with human umbilical vein endothelial cells (HUVECs) which showed a high expression level of Nrf2. A short hairpin RNA (shRNA) and Sulforaphane (SFN) affecting the expression of Nrf2 were used to treat HUVECs. The results showed that Nrf2 could promote the tube formation of HUVECs. Nrf2 also exerts a protective effect against HUVECs, which is achieved by promoting NQO1 expression to stabilize ROS levels. Furthermore, autophagy activation significantly promotes angiogenesis in the spinal echinococcosis model (SEM) as a result of Nrf2 regulation of oxidative stress. These results suggest that the ROS/Nrf2/autophagy axis can induce angiogenesis and may be a potential target for the treatment of spinal cystic echinococcosis.
Collapse
Affiliation(s)
- Qian Ren
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yimin Dong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430101, China
| | - Yiping Huang
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Jun Xiao
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yibo Ma
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yaqing Liu
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Haohao Sun
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Yi Dai
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China
| | - Chenhui Shi
- Laboratory for Bone Cystic Echinococcosis Research, Orthopaedic Centre, The First Affiliated Hospital of Shihezi University, Shihezi City Xinjiang Uygur Autonomous Region, 832000, China.
| | - Sibo Wang
- Department of Spine Surgery, Xi'an Jiao Tong University Affiliated HongHui Hospital, Xi'an 710054, China.
| |
Collapse
|
5
|
Duan Y, Zhang W, Ouyang Y, Yang Q, Zhang Q, Zhao S, Chen C, Xu T, Zhang Q, Ran H, Liu H. Proton Sponge Nanocomposites for Synergistic Tumor Elimination via Autophagy Inhibition-Promoted Cell Apoptosis and Macrophage Repolarization-Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:17285-17299. [PMID: 38539044 DOI: 10.1021/acsami.4c01451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Cytoprotective autophagy and an immunosuppressive tumor microenvironment (TME) are two positive promoters for tumor proliferation and metastasis that severely hinder therapeutic efficacy. Inhibiting autophagy and reconstructing TME toward macrophage activation simultaneously are of great promise for effective tumor elimination, yet are still a huge challenge. Herein, a kind of dendrimer-based proton sponge nanocomposites was designed and constructed for tumor chemo/chemodynamic/immunotherapy through autophagy inhibition-promoted cell apoptosis and macrophage repolarization-enhanced immune response. These obtained nanocomposites contain a proton sponge G5AcP dendrimer, a Fenton-like agent Cu(II), and chemical drug doxorubicin (DOX). When accumulated in tumor regions, G5AcP can act as an immunomodulator to realize deacidification-promoted macrophage repolarization toward antitumoral type, which then secretes inflammatory cytokines to activate T cells. They also regulate intracellular lysosomal pH to inhibit cytoprotective autophagy. The released Cu(II) and DOX can induce aggravated damage through a Fenton-like reaction and chemotherapeutic effect in this autophagy-inhibition condition. Tumor-associated antigens are released from these dying tumor cells to promote the maturity of dendritic cells, further activating T cells. Effective tumor elimination can be achieved by this dendrimer-based therapeutic strategy, providing significant guidance for the design of a promising antitumor nanomedicine.
Collapse
Affiliation(s)
- Yifan Duan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yi Ouyang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qiang Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qiuye Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Sheng Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Chunmei Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Ting Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qun Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hui Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
6
|
Elgendy SM, Zaher DM, Sarg NH, Abu Jayab NN, Alhamad DW, Al-Tel TH, Omar HA. Autophagy inhibition potentiates energy restriction-induced cell death in hepatocellular carcinoma cells. IUBMB Life 2024. [PMID: 38497226 DOI: 10.1002/iub.2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/18/2024] [Indexed: 03/19/2024]
Abstract
Hepatocellular carcinoma (HCC) significantly contributes to cancer-related mortality due to the limited response of HCC to current anticancer therapies, thereby necessitating more effective treatment approaches. Energy restriction mimetic agents (ERMAs) have emerged as potential therapies in targeting the Warburg effect, a unique metabolic process in cancer cells. However, ERMAs exhibit limited efficacy when used as monotherapy. Additionally, ERMAs have been found to induce autophagy in cancer cells. The role of autophagy in cancer survival remains a subject of debate. Thus, it is crucial to ascertain whether ERMA-induced autophagy is a mechanism for cell survival or cell death in HCC. Our study aims to investigate the effect of autophagy inhibition on the survival of HCC cells treated with ERMAs while also examining the potential of combining an autophagy inhibitor such as spautin-1 with ERMAs to enhance HCC cell death. Our results suggest a cytoprotective role for ERMA-induced autophagy in HCC cells, as combining the autophagy inhibitor spautin-1 with ERMAs effectively suppressed ERMA-induced autophagy and synergistically enhanced their antitumor activity. The treatment combination promoted HCC death through apoptosis, cell cycle arrest, and inhibition of AKT and ERK activation, which are known to play a key role in cellular proliferation. Collectively, our findings highlight a potential strategy to combat HCC by combining energy restriction with autophagy inhibition.
Collapse
Affiliation(s)
- Sara M Elgendy
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Dana M Zaher
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Nadin H Sarg
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Nour N Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Dima W Alhamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Taleb H Al-Tel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Hany A Omar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
7
|
Guo J, Lei Y, Liu L, Wen Z, Zhang B, Fang J, Liang G, Guo Q, Peng J. MHY1485 promotes adriamycin sensitivity in HepG2 cells by inhibiting autophagy. Funct Integr Genomics 2024; 24:22. [PMID: 38306005 DOI: 10.1007/s10142-024-01304-3] [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: 11/14/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
MHY1485 is an mTOR activator that inhibits the autophagy process by inhibiting the fusion between autophagosomes and lysosomes. This study aimed to explore the role and mechanism of MHY1485 in hepatocellular carcinoma (HCC) and to provide an in-depth understanding of the mechanisms of autophagy regulation in relation to adriamycin (ADM) resistance, as well as the development of a molecularly targeted autophagy-modulating approach. Here, ADM was used to treat HepG2 cells and construct an ADM-resistant cell model. The HepG2/ADM cell line and HepG2 cells were treated with MHY1485 and ADM, respectively, and the proliferation and apoptosis of HCC cells were detected using CCK8, clone formation, flow cytometry, and 5-ethynyl-2'-deoxyuridine staining assays. Ki-67, mTOR phosphorylation, and LC3A expression were detected by IF staining; the expression or phosphorylation levels of autophagy-related proteins (i.e., GLUT1, PGI, PFK, END, and MTHFD2) and apoptosis-related proteins (caspase-3, caspase-8, and caspase-9) were detected by qPCR and western blotting. The number of autophagosomes was determined by monodansylcadaverine staining. Our results showed that MHY1485 can inhibit the proliferation and growth of liver cancer cells, and that MHY1485 combined with ADM can effectively inhibit the tolerance of HepG2/ADM cells to ADM and enhance the efficacy of ADM. The results of the detection of the autophagy-related protein LC3A also indicated that MHY1485 activates mTOR and can affect the phosphorylation level of ULK1, inhibit autophagy, and enhance the sensitivity of liver cancer cells to adriamycin. In summary, MHY1485 can enhance the sensitivity of adriamycin-resistant cells to adriamycin by activating mTOR and blocking the autophagy process in cells; therefore, mTOR may become a potential target for the treatment of liver cancer.
Collapse
Affiliation(s)
- Jingfeng Guo
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China.
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China.
| | - Yingying Lei
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Liwei Liu
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Zhenzhen Wen
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Bo Zhang
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Jincun Fang
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Guohui Liang
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Qikun Guo
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| | - Jing Peng
- Department of Medical Oncology, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Panyu Maternal and Child Care Service Center of Guangzhou, Guangzhou, China
| |
Collapse
|
8
|
Li Y, Guo Y, Zhang K, Zhu R, Chen X, Zhang Z, Yang W. Cell Death Pathway Regulation by Functional Nanomedicines for Robust Antitumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306580. [PMID: 37984863 PMCID: PMC10797449 DOI: 10.1002/advs.202306580] [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: 09/12/2023] [Revised: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Cancer immunotherapy has become a mainstream cancer treatment over traditional therapeutic modes. Cancer cells can undergo programmed cell death including ferroptosis, pyroptosis, autophagy, necroptosis, apoptosis and cuproptosis which are find to have intrinsic relationships with host antitumor immune response. However, direct use of cell death inducers or regulators may bring about severe side effects that can also be rapidly excreted and degraded with low therapeutic efficacy. Nanomaterials are able to carry them for long circulation time, high tumor accumulation and controlled release to achieve satisfactory therapeutic effect. Nowadays, a large number of studies have focused on nanomedicines-based strategies through modulating cell death modalities to potentiate antitumor immunity. Herein, immune cell types and their function are first summarized, and state-of-the-art research progresses in nanomedicines mediated cell death pathways (e.g., ferroptosis, pyroptosis, autophagy, necroptosis, apoptosis and cuproptosis) with immune response provocation are highlighted. Subsequently, the conclusion and outlook of potential research focus are discussed.
Collapse
Affiliation(s)
- Yongjuan Li
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
- Medical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouHenan450001China
- The center of Infection and ImmunityAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450001China
| | - Yichen Guo
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
| | - Kaixin Zhang
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
| | - Rongrong Zhu
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, SurgeryChemical and Biomolecular Engineering, and Biomedical EngineeringYong Loo Lin School of Medicine and Faculty of EngineeringNational University of SingaporeSingapore119074Singapore
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingapore117599Singapore
- Nanomedicine Translational Research ProgramNUS Center for NanomedicineYong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Zhenzhong Zhang
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
| | - Weijing Yang
- School of Pharmaceutical SciencesHenan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhouHenan450001China
| |
Collapse
|
9
|
Sun Y, Lian T, Huang Q, Chang Y, Li Y, Guo X, Kong W, Yang Y, Zhang K, Wang P, Wang X. Nanomedicine-mediated regulated cell death in cancer immunotherapy. J Control Release 2023; 364:174-194. [PMID: 37871752 DOI: 10.1016/j.jconrel.2023.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Immunotherapy has attracted widespread attention in cancer treatment and has achieved considerable success in the clinical treatment of some tumors, but it has a low response rate in most tumors. To achieve sufficient activation of the immune response, significant efforts using nanotechnology have been made to enhance cancer immune response. In recent years, the induction of various regulated cell death (RCD) has emerged as a potential antitumor immuno-strategy, including processes related to apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis. In particular, damage-associated molecular patterns (DAMPs) released from the damaged membrane of dying cells act as in situ adjuvants to trigger antigen-specific immune responses by the exposure of an increased antigenicity. Thus, RCD-based immunotherapy offers a new approach for enhancing cancer treatment efficacy. Furthermore, incorporation with multimodal auxiliary therapies in cell death-based immunotherapy can trigger stronger immune responses, resulting in more efficient therapeutic outcome. This review discusses different RCD modalities and summarizes recent nanotechnology-mediated RCDs in cancer immunotherapy.
Collapse
Affiliation(s)
- Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; The Xi'an key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Ting Lian
- Research Center for Prevention and Treatment of Respiratory Disease, School of Clinical Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Qichao Huang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yawei Chang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaoyu Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Weirong Kong
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yifang Yang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
10
|
Demir AB, Baris E, Kaner UB, Alotaibi H, Atabey N, Koc A. Toll-interacting protein may affect doxorubicin resistance in hepatocellular carcinoma cell lines. Mol Biol Rep 2023; 50:8551-8563. [PMID: 37644370 DOI: 10.1007/s11033-023-08737-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Liver cancer is the third leading cause of cancer-related deaths worldwide, and hepatocellular carcinoma (HCC) is the most common type of liver cancer. Transarterial interventions are among the chemotherapeutic approaches used in hardly operable regions prior to transplantation, and in electrochemotherapy, where doxorubicin is used. However, the efficacy of treatment is affected by resistance mechanisms. Previously, we showed that overexpression of the CUE5 gene results in doxorubicin resistance in Saccharomyces cerevisiae (S. cerevisiae). In this study, the effect of Toll-interacting protein (TOLLIP), the human ortholog of CUE5, on doxorubicin resistance was evaluated in HCC cells to identify its possible role in increasing the efficacy of transarterial interventions. METHODS AND RESULTS The NIH Gene Expression Omnibus (GEO) and Oncomine datasets were analyzed for HCC cell lines with relatively low and high TOLLIP expression, and SNU449 and Hep3B cell lines were chosen, respectively. TOLLIP expression was increased by plasmid transfection and decreased by TOLLIP-siRNA in both cell lines and evaluated by RT-PCR and ELISA. Cell proliferation and viability were examined using xCELLigence and MTT assays after doxorubicin treatment, and growth inhibitory 50 (GI 50) concentrations were evaluated. Doxorubicin GI 50 concentrations decreased approximately 2-folds in both cell lines upon silencing TOLLIP after 48 h of drug treatment. CONCLUSIONS Our results showed for the first time that silencing TOLLIP in hepatocellular carcinoma cells may help sensitize these cells to doxorubicin and increase the efficacy of chemotherapeutic regimens where doxorubicin is used.
Collapse
Affiliation(s)
- Ayse Banu Demir
- Faculty of Medicine, Department of Medical Biology, Izmir University of Economics, Sakarya Street, No:156, Balcova, Izmir, 35330, Turkey.
| | - Elif Baris
- Faculty of Medicine, Department of Medical Pharmacology, Izmir University of Economics, Izmir, Turkey
| | - Umay Bengi Kaner
- Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
| | - Hani Alotaibi
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University Health Campus, Izmir, Turkey
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Nese Atabey
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Faculty of Medicine, Department of Medical Biology & Galen Research Center, Izmir Tinaztepe University, Izmir, Turkey
| | - Ahmet Koc
- Faculty of Medicine, Department of Medical Genetics, Inonu University, Malatya, Turkey
| |
Collapse
|
11
|
Xing L, Tang Y, Li L, Tao X. ROS in hepatocellular carcinoma: What we know. Arch Biochem Biophys 2023:109699. [PMID: 37499994 DOI: 10.1016/j.abb.2023.109699] [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: 05/25/2023] [Revised: 07/07/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Hepatocellular carcinoma (HCC), which is a primary liver cancer subtype, has a poor prognosis due to its high degree of malignancy. The lack of early diagnosis makes systemic therapy the only hope for HCC patients with advanced disease; however, resistance to drugs is a major obstacle. In recent years, targeted molecular therapy has gained popularity as a potential treatment for HCC. An increase in reactive oxygen species (ROS), which are cancer markers and a potential target for HCC therapy, can both promote and inhibit the disease. At present, many studies have examined targeted regulation of ROS in the treatment of HCC. Here, we reviewed the latest drugs that are still in the experimental stage, including nanocarrier drugs, exosome drugs, antibody drugs, aptamer drugs and polysaccharide drugs, to provide new hope for the clinical treatment of HCC patients.
Collapse
Affiliation(s)
- Lin Xing
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; School of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yuting Tang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; School of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Lu Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
12
|
Qiu Y, Wang X, Sun Y, Du YE, Yin G, Luo H, Wen S, Lang L, Liu M, Tang X. TCF12 regulates exosome release from epirubicin-treated CAFs to promote ER+ breast cancer cell chemoresistance. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166727. [PMID: 37137433 DOI: 10.1016/j.bbadis.2023.166727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are the predominant stromal cells in the microenvironment and play important roles in tumor progression, including chemoresistance. However, the response of CAFs to chemotherapeutics and their effects on chemotherapeutic outcomes are largely unknown. In this study, we showed that epirubicin (EPI) treatment triggered ROS which initiated autophagy in CAFs, TCF12 inhibited autophagy flux and further promoted exosome secretion. Inhibition of EPI-induced reactive oxygen species (ROS) production with N-acetyl-L-cysteine (NAC) or suppression of autophagic initiation with short interfering RNA (siRNA) against ATG5 blunted exosome release from CAFs. Furthermore, exosome secreted from EPI-treated CAFs not only prevented ROS accumulation in CAFs but also upregulated the CXCR4 and c-Myc protein levels in recipient ER+ breast cancer cells, thus promoting EPI resistance of tumor cells. Together, the current study provides novel insights into the role of stressed CAFs in promoting tumor chemoresistance and reveal a new function of TCF12 in regulating autophagy impairment and exosome release.
Collapse
Affiliation(s)
- Yuxiang Qiu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xing Wang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Yan-E Du
- Department of Laboratory Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyang Wen
- Department of Laboratory Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Lei Lang
- Department of Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xi Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
13
|
Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S, Mansouri K. Autophagy: A challengeable paradox in cancer treatment. Cancer Med 2023. [PMID: 36760166 DOI: 10.1002/cam4.5577] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Autophagy is an intracellular degradation pathway conserved in all eukaryotes from yeast to humans. This process plays a quality-control role by destroying harmful cellular components under normal conditions, maintaining cell survival, and establishing cellular adaptation under stressful conditions. Hence, there are various studies indicating dysfunctional autophagy as a factor involved in the development and progression of various human diseases, including cancer. In addition, the importance of autophagy in the development of cancer has been highlighted by paradoxical roles, as a cytoprotective and cytotoxic mechanism. Despite extensive research in the field of cancer, there are many questions and challenges about the roles and effects suggested for autophagy in cancer treatment. The aim of this study was to provide an overview of the paradoxical roles of autophagy in different tumors and related cancer treatment options. METHODS In this study, to find articles, a search was made in PubMed and Google scholar databases with the keywords Autophagy, Autophagy in Cancer Management, and Drug Design. RESULTS According to the investigation, some studies suggest that several advanced cancers are dependent on autophagy for cell survival, so when cancer cells are exposed to therapy, autophagy is induced and suppresses the anti-cancer effects of therapeutic agents and also results in cell resistance. However, enhanced autophagy from using anti-cancer drugs causes autophagy-mediated cell death in several cancers. Because autophagy also plays roles in both tumor suppression and promotion further research is needed to determine the precise mechanism of this process in cancer treatment. CONCLUSION We concluded in this article, autophagy manipulation may either promote or hinder the growth and development of cancer according to the origin of the cancer cells, the type of cancer, and the behavior of the cancer cells exposed to treatment. Thus, before starting treatment it is necessary to determine the basal levels of autophagy in various cancers.
Collapse
Affiliation(s)
- Farnaz Ahmadi-Dehlaghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Biology, Payame Noor University, Tehran, Iran
| | - Parisa Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elahe Valipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sarah Kiani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
14
|
Zhang T, Gan Y, Zhu S. Association between autophagy and acute pancreatitis. Front Genet 2023; 14:998035. [PMID: 36793898 PMCID: PMC9923090 DOI: 10.3389/fgene.2023.998035] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Autophagy pathway involves maintaining intracellular homeostasis by regulating the degradation of cytoplasmic components. Disfunction of autophagic process has been confirmed to be critical mechanism in many diseases, including cancer, inflammation, infection, degeneration and metabolic disorders. Recent studies have shown that autophagy is one of the early events in acute pancreatitis. Impaired autophagy promotes the abnormal activation of zymogen granules and results in apoptosis and necrosis of exocrine pancreas. Furthermore, multiple signal paths involve progression of acute pancreatitis by regulating autophagy pathway. This article provides a comprehensive review of the recent advances in epigenetic regulation of autophagy and the role of autophagy in acute pancreatitis.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, China,Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Yu Gan, ; Shuai Zhu,
| | - Shuai Zhu
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, China,Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China,*Correspondence: Yu Gan, ; Shuai Zhu,
| |
Collapse
|
15
|
Zhang Y, Li J, Pu K. Recent advances in dual- and multi-responsive nanomedicines for precision cancer therapy. Biomaterials 2022; 291:121906. [DOI: 10.1016/j.biomaterials.2022.121906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
|
16
|
Yao H, Gong X, Geng M, Duan S, Qiao P, Sun F, Zhu Z, Du B. Cascade nanozymes based on the "butterfly effect" for enhanced starvation therapy through the regulation of autophagy. Biomater Sci 2022; 10:4008-4022. [PMID: 35726640 DOI: 10.1039/d2bm00595f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although tumor starvation therapy has been proven to be an excellent method for tumor therapy, its efficiency may be weakened by autophagy, a self-protection mechanism exerted by tumors under starvation stress. Interestingly, over-activated autophagy not only improves the efficacy of starvation therapy, but also induces autophagic death. Herein, we report cascade nanozymes for enhanced starvation therapy by inducing over-activated autophagy. First, glucose oxidase (GOx) modified metal-organic frameworks (NH2-MIL88, MOF) were constructed (MOF-GOx). After loading with curcumin (Cur), Cur@MOF-GOx was further decorated with tumor-targeting hyaluronic acid (HA) to obtain Cur@MOF-GOx/HA nanozymes. GOx can catalyze glucose into H2O2 and gluconic acid, which not only leads to tumor starvation, but also provides reactants for the Fenton reaction mediated by the MOF to generate hydroxyl radicals (˙OH) for chemo-dynamic therapy. Most importantly, protective autophagy caused by tumor starvation can be over-activated by Cur to convert autophagy from pro-survival to pro-death, realizing augmented anticancer therapy efficacy. With these cascade reactions, the synergistic action of starvation, autophagy and chemo-dynamic therapy was realized. Generally, the introduction of Cur@MOF-GOx/HA into tumor cells leads to a "butterfly effect", which induces enhanced starvation therapy through subsequent autophagic cell death to completely break the self-protective mechanism of cancer cells, and generate ˙OH for chemo-dynamic therapy. Precise design allows for the use of cascade nanozymes to realize efficient cancer treatment and restrain metastasis.
Collapse
Affiliation(s)
- Hanchun Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China.,Collaborative Innovation Center of Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, China
| | - Xiaobao Gong
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Meilin Geng
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Songchao Duan
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Pan Qiao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Fangfang Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Zhihui Zhu
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Du
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China.,Collaborative Innovation Center of Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, China
| |
Collapse
|
17
|
Wang R, Fu Y, Yao M, Cui X, Zhao Y, Lu X, Li Y, Lin Y, He S. The HN1/HMGB1 axis promotes the proliferation and metastasis of hepatocellular carcinoma and attenuates the chemosensitivity to oxaliplatin. FEBS J 2022; 289:6400-6419. [PMID: 35596723 DOI: 10.1111/febs.16531] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/21/2022] [Accepted: 05/19/2022] [Indexed: 11/26/2022]
Abstract
Hematological and neurological expressed 1 (HN1) is closely associated with the proliferation and metastasis of various tumors. However, the physiological functions and clinical significance of HN1 in hepatocellular carcinoma (HCC) remain indistinct. In this study, we investigated the role of HN1 in the pathogenesis of HCC and the underlying mechanism using clinical data from HCC patients, in vitro experiments utilizing HCC cell lines and in vivo animal models. We demonstrated that the overexpressed HN1 in HCC was correlated with patients' adverse outcomes. The gain and loss of function experiments indicated that HN1 could promote the proliferation, migration, and invasion of HCC cells in vitro. Furthermore, we found that HN1 knockdown sensitized HCC cells to oxaliplatin. Mechanically, HN1 prevented HMGB1 protein from ubiquitination and degradation via the autophagy-lysosome pathway, which was related to the interaction between HN1 protein and TRIM28 protein. In the nucleus, the downregulation of HMGB1 followed by HN1 knockdown resulted in increased DNA damage and cell death in the oxaliplatin-treated HCC cells. In the cytoplasm, HN1 regulated autophagy via HMGB1. Furthermore, HN1 knockdown in combination with HMGB1 overexpression restored the aggressive phenotypes of HCC cells and the sensitivity of these cells to oxaliplatin. HN1 knockdown inhibited the tumor growth and metastasis, and promoted the anticancer efficiency of oxaliplatin in vivo. In conclusion, our data suggest that the HN1/HMGB1 axis plays an important role in the development/progression and chemotherapy of HCC. Our findings indicate that the HN1/HMGB1 axis may be a promising therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Ruhua Wang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yunong Fu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Menglin Yao
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xiaomeng Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xinlan Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yarui Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
18
|
Gao Z, Chen JF, Li XG, Shi YH, Tang Z, Liu WR, Zhang X, Huang A, Luo XM, Gao Q, Shi GM, Ke AW, Zhou J, Fan J, Fu XT, Ding ZB. KRAS acting through ERK signaling stabilizes PD-L1 via inhibiting autophagy pathway in intrahepatic cholangiocarcinoma. Cancer Cell Int 2022; 22:128. [PMID: 35305624 PMCID: PMC8933925 DOI: 10.1186/s12935-022-02550-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/13/2022] [Indexed: 12/23/2022] Open
Abstract
Background While the correlation between PD-L1 expression and KRAS mutation has been previously reported in other solid tumors such as non-small cell lung cancer (NSCLC), whether PD-L1 can be modulated by ERK signaling downstream of KRAS in intrahepatic cholangiocarcinoma (iCCA) and the underlying molecular regulatory mechanism remain unclear. Methods The expression of ERK, p-ERK, PD-L1 and autophagy markers following KRAS knockdown or Ras/Raf/MEK/ERK signaling inhibitors treatment was examined in two human iCCA cell lines (HuCCT1 and RBE) using western blotting and immunofluorescence. Both pharmacological autophagy inhibitors and short-interfering RNA against ATG7 were applied to inhibit autophagy. The apoptosis rates of iCCA cell lines were detected by flow cytometry and CCK-8 after co-culturing with CD3/CD28-activated human CD8+ T lymphocytes. Immunohistochemistry was applied to detect the correlation of ERK, p-ERK and PD-L1 in 92 iCCA tissues. Results The present study demonstrated that the PD-L1 expression level was distinctly reduced in KRAS-mutated iCCA cell lines when ERK signaling was inhibited and ERK phosphorylation levels were lowered. The positive association between p-ERK and PD-L1 was also verified in 92 iCCA tissue samples. Moreover, ERK inhibition induced autophagy activation. Both inhibiting autophagy via autophagy inhibitors and genetically silencing the ATG7 expression partially reversed the reduced PD-L1 expression caused by ERK inhibition. In addition, ERK-mediated down-regulation of PD-L1 via autophagy pathways induced the apoptosis of iCCA cells when co-cultured with CD3/CD28-activated human CD8+ T lymphocytes in vitro. Conclusions Our results suggest that ERK signaling inhibition contributes to the reduction of PD-L1 expression through the autophagy pathway in iCCA. As a supplement to anti-PD-1/PD-L1 immunotherapy, ERK-targeted therapy may serve as a potentially novel treatment strategy for human KRAS-mutated iCCA.
Collapse
|
19
|
Yuan M, Shong K, Li X, Ashraf S, Shi M, Kim W, Nielsen J, Turkez H, Shoaie S, Uhlen M, Zhang C, Mardinoglu A. A Gene Co-Expression Network-Based Drug Repositioning Approach Identifies Candidates for Treatment of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14061573. [PMID: 35326724 PMCID: PMC8946504 DOI: 10.3390/cancers14061573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common malignancy of liver cancer. However, treatment of HCC is still severely limited due to limitation of drug therapy. We aimed to screen more possible target genes and candidate drugs for HCC, exploring the possibility of drug treatments from systems biological perspective. We identified ten candidate target genes, which are hub genes in HCC co-expression networks, which also possess significant prognostic value in two independent HCC cohorts. The rationality of these target genes was well demonstrated through variety analyses of patient expression profiles. We then screened candidate drugs for target genes and finally identified withaferin-a and mitoxantrone as the candidate drug for HCC treatment. The drug effectiveness was validated in in vitro model and computational analysis, providing more evidence for our drug repositioning method and results. Abstract Hepatocellular carcinoma (HCC) is a malignant liver cancer that continues to increase deaths worldwide owing to limited therapies and treatments. Computational drug repurposing is a promising strategy to discover potential indications of existing drugs. In this study, we present a systematic drug repositioning method based on comprehensive integration of molecular signatures in liver cancer tissue and cell lines. First, we identify robust prognostic genes and two gene co-expression modules enriched in unfavorable prognostic genes based on two independent HCC cohorts, which showed great consistency in functional and network topology. Then, we screen 10 genes as potential target genes for HCC on the bias of network topology analysis in these two modules. Further, we perform a drug repositioning method by integrating the shRNA and drug perturbation of liver cancer cell lines and identifying potential drugs for every target gene. Finally, we evaluate the effects of the candidate drugs through an in vitro model and observe that two identified drugs inhibited the protein levels of their corresponding target genes and cell migration, also showing great binding affinity in protein docking analysis. Our study demonstrates the usefulness and efficiency of network-based drug repositioning approach to discover potential drugs for cancer treatment and precision medicine approach.
Collapse
Affiliation(s)
- Meng Yuan
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
| | - Koeun Shong
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
| | - Xiangyu Li
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
- Bash Biotech Inc., 600 West Broadway, Suite 700, San Diego, CA 92101, USA
| | - Sajda Ashraf
- Heka Lab, Camlik Mah. Hearty, Sk. No:4 Heka Human Plaza Umraniye, Istanbul 34774, Turkey;
| | - Mengnan Shi
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
| | - Woonghee Kim
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden;
- BioInnovation Institute, DK-2200 Copenhagen, Denmark
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey;
| | - Saeed Shoaie
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| | - Mathias Uhlen
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
| | - Cheng Zhang
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
- Key Laboratory of Advanced Drug Preparation Technologies, School of Pharmaceutical Sciences, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (C.Z.); (A.M.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (M.Y.); (K.S.); (X.L.); (M.S.); (W.K.); (S.S.); (M.U.)
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
- Correspondence: (C.Z.); (A.M.)
| |
Collapse
|
20
|
Widjaya AS, Liu Y, Yang Y, Yin W, Liang J, Jiang Y. Tumor-permeable smart liposomes by modulating the tumor microenvironment to improve the chemotherapy. J Control Release 2022; 344:62-79. [PMID: 35182612 DOI: 10.1016/j.jconrel.2022.02.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022]
Abstract
Low levels of accumulation and permeability in tumors are two primary reasons for the limited efficacy of conventional antineoplastic nanodrugs. In the present study, based on an original corosolic acid liposome (CALP) carrier with the functions of cell penetration, tumor permeability and anti-inflammation developed by our previous work, a versatile PTX/CALP was achieved by CALP loading paclitaxel (PTX). Compared to conventional PTX liposomes (PTX/LP) prepared by cholesterol and phospholipid, PTX/CALP exhibited extremely increasing cellular uptake and cytotoxicity in vitro, and in vivo enhancing the accumulation and permeability of tumor, thus significantly improving the antitumor efficacy. Further evidence indicated that PTX/CALP conspicuously promoted the recruitment of CD8+ T cells as well as reduced the infiltration of regulatory T cells and M2 macrophages into tumor by inducing enhanced immunogenic cell death (ICD) and down-regulating the inflammation level. Therefore, the improvement of efficacy was also attributed to the superiorities of PTX/CALP in modulating the inflammatory and immunosuppressive tumor microenvironment. Overall, the smart PTX liposomes based on the multi-functional CALP carrier without any modification could overcome the harsh tumor biological barriers, enhance the induction of ICD and then achieve satisfactory efficacy, suggesting its promising potentials in industrial transfer and clinical application.
Collapse
Affiliation(s)
- Andy Samuel Widjaya
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunhu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yueying Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weiwei Yin
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianying Liang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
21
|
Kim MJ, Choi B, Kim JY, Min Y, Kwon DH, Son J, Lee JS, Lee JS, Chun E, Lee KY. USP8 regulates liver cancer progression via the inhibition of TRAF6-mediated signal for NF-κB activation and autophagy induction by TLR4. Transl Oncol 2022; 15:101250. [PMID: 34688043 PMCID: PMC8546492 DOI: 10.1016/j.tranon.2021.101250] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Herein, we aimed to elucidate the molecular and cellular mechanism in which ubiquitin-specific protease 8 (USP8) is implicated in liver cancer progression via TRAF6-mediated signal. USP8 induces the deubiquitination of TRAF6, TAB2, TAK1, p62, and BECN1, which are pivotal roles for NF-κB activation and autophagy induction. Notably, the LIHC patient with low USP8 mRNA expression showed markedly shorter survival time, whereas there was no significant difference in the other 18-human cancers. Importantly, the TCGA data analysis on LIHC and transcriptome analysis on the USP8 knockout (USP8KO) SK-HEP-1 cells revealed a significant correlation between USP8 and TRAF6, TAB2, TAK1, p62, and BECN1, and enhanced NF-κB-dependent and autophagy-related cancer progression/metastasis-related genes in response to LPS stimulation. Furthermore, USP8KO SK-HEP-1 cells showed an increase in cancer migration and invasion by TLR4 stimulation, and a marked increase of tumorigenicity and metastasis in xenografted NSG mice. The results demonstrate that USP8 is negatively implicated in the LIHC progression through the regulation of TRAF6-mediated signal for the activation of NF-κB activation and autophagy induction. Our findings provide useful insight into the LIHC pathogenesis of cancer progression.
Collapse
Affiliation(s)
- Mi-Jeong Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Bongkum Choi
- Department of Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Ji Young Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Yoon Min
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Do Hee Kwon
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juhee Son
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Ji Su Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Joo Sang Lee
- Department of Precision medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Eunyoung Chun
- CHA Vaccine Institute, 560 Dunchon-daero, Jungwon-gu, Seongnam-si, Gyeonggi-do 13230, Republic of Korea.
| | - Ki-Young Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Shi Y, Lin G, Zheng H, Mu D, Chen H, Lu Z, He P, Zhang Y, Liu C, Lin Z, Liu G. Biomimetic nanoparticles blocking autophagy for enhanced chemotherapy and metastasis inhibition via reversing focal adhesion disassembly. J Nanobiotechnology 2021; 19:447. [PMID: 34952594 PMCID: PMC8710033 DOI: 10.1186/s12951-021-01189-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Autophagy is a conserved catabolic process, which plays an important role in regulating tumor cell motility and degrading protein aggregates. Chemotherapy-induced autophagy may lead to tumor distant metastasis and even chemo-insensitivity in the therapy of hepatocellular carcinoma (HCC). Therefore, a vast majority of HCC cases do not produce a significant response to monotherapy with autophagy inhibitors. RESULTS In this work, we developed a biomimetic nanoformulation (TH-NP) co-encapsulating Oxaliplatin (OXA)/hydroxychloroquine (HCQ, an autophagy inhibitor) to execute targeted autophagy inhibition, reduce tumor cell migration and invasion in vitro and attenuate metastasis in vivo. The tumor cell-specific ligand TRAIL was bioengineered to be stably expressed on HUVECs and the resultant membrane vesicles were wrapped on OXA/HCQ-loaded PLGA nanocores. Especially, TH-NPs could significantly improve OXA and HCQ effective concentration by approximately 21 and 13 times in tumor tissues compared to the free mixture of HCQ/OXA. Moreover, the tumor-targeting TH-NPs released HCQ alkalized the acidic lysosomes and inhibited the fusion of autophagosomes and lysosomes, leading to effective blockade of autophagic flux. In short, the system largely improved chemotherapeutic performance of OXA on subcutaneous and orthotopic HCC mice models. Importantly, TH-NPs also exhibited the most effective inhibition of tumor metastasis in orthotopic HCCLM3 models, and in the HepG2, Huh-7 or HCCLM3 metastatic mice models. Finally, we illustrated the enhanced metastasis inhibition was attributed to the blockade or reverse of the autophagy-mediated degradation of focal adhesions (FAs) including E-cadherin and paxillin. CONCLUSIONS TH-NPs can perform an enhanced chemotherapy and antimetastatic effect, and may represent a promising strategy for HCC therapy in clinics.
Collapse
Affiliation(s)
- Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361004, China
| | - Gan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Huili Zheng
- Department of Anesthesiology, Zhongshan Hospital of Xiamen University, Xiamen, 361004, China
| | - Dan Mu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,Amoy Hopeful Biotechnology Co., Ltd., Xiamen, 361027, China
| | - Hu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,Amoy Hopeful Biotechnology Co., Ltd., Xiamen, 361027, China
| | - Zhixiang Lu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Pan He
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Zhongning Lin
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China. .,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361004, China.
| |
Collapse
|
23
|
Ma L, Xu A, Kang L, Cong R, Fan Z, Zhu X, Huo N, Liu W, Xue C, Ji Q, Li W, Chu Z, Kang X, Wang Y, Sun Z, Han Y, Liu H, Gao X, Han J, You H, Zhao C, Xu X. LSD1-Demethylated LINC01134 Confers Oxaliplatin Resistance Through SP1-Induced p62 Transcription in HCC. Hepatology 2021; 74:3213-3234. [PMID: 34322883 DOI: 10.1002/hep.32079] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Oxaliplatin (OXA) is one of the most common chemotherapeutics in advanced hepatocellular carcinoma (HCC), the resistance of which poses a big challenge. Long noncoding RNAs (lncRNAs) play vital roles in chemoresistance. Therefore, elucidating the underlying mechanisms and identifying predictive lncRNAs for OXA resistance is needed urgently. METHODS RNA sequencing (RNA-seq) and fluorescence in situ hybridization (FISH) were used to investigate the OXA-resistant (OXA-R) lncRNAs. Survival analysis was performed to determine the clinical significance of homo sapiens long intergenic non-protein-coding RNA 1134 (LINC01134) and p62 expression. Luciferase, RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP), and chromatin isolation by RNA purification (ChIRP) assays were used to explore the mechanisms by which LINC01134 regulates p62 expression. The effects of LINC01134/SP1/p62 axis on OXA resistance were evaluated using cell viability, apoptosis, and mitochondrial function and morphology analysis. Xenografts were used to estimate the in vivo regulation of OXA resistance by LINC01134/SP1/p62 axis. ChIP, cell viability, and xenograft assays were used to identify the demethylase for LINC01134 up-regulation in OXA resistance. RESULTS LINC01134 was identified as one of the most up-regulated lncRNAs in OXA-R cells. Higher LINC01134 expression predicted poorer OXA therapeutic efficacy. LINC01134 activates anti-oxidative pathway through p62 by recruiting transcription factor SP1 to the p62 promoter. The LINC01134/SP1/p62 axis regulates OXA resistance by altering cell viability, apoptosis, and mitochondrial homeostasis both in vitro and in vivo. Furthermore, the demethylase, lysine specific demethylase 1 (LSD1) was responsible for LINC01134 up-regulation in OXA-R cells. In patients with HCC, LINC01134 expression was positively correlated with p62 and LSD1 expressions, whereas SP1 expression positively correlated with p62 expression. CONCLUSIONS LSD1/LINC01134/SP1/p62 axis is critical for OXA resistance in HCC. Evaluating LINC01134 expression in HCC will be effective in predicting OXA efficacy. In treatment-naive patients, targeting the LINC01134/SP1/p62 axis may be a promising strategy to overcome OXA chemoresistance.
Collapse
Affiliation(s)
- Luyuan Ma
- Department of Infectious Diseases, the Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - An Xu
- Department of Oncology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Rui Cong
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Zhongyi Fan
- Department of Oncology and Hematology, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Xiang Zhu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Nan Huo
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wenpeng Liu
- Department of Infectious Diseases, the Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunyuan Xue
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Quanbo Ji
- Department of Orthopedics, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenchao Li
- Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhong Chu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaofeng Kang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yadong Wang
- Department of Infectious Diseases, the Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhijia Sun
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yuchen Han
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Hanxiao Liu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Juqiang Han
- Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hua You
- Department of Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Caiyan Zhao
- Department of Infectious Diseases, the Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaojie Xu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
24
|
Sun F, Liu Y, Gong T, Pan Q, Xiang T, Zhao J, Tang Y, Chen H, Han Y, Song M, Huang Y, Li H, Chen Y, Yang C, Yang J, Wang Q, Li Y, He J, Weng D, Peng R, Xia J. Inhibition of DTYMK significantly restrains the growth of HCC and increases sensitivity to oxaliplatin. Cell Death Dis 2021; 12:1093. [PMID: 34795209 PMCID: PMC8602592 DOI: 10.1038/s41419-021-04375-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022]
Abstract
Most patients with hepatocellular carcinoma (HCC) are in the middle or advanced stage at the time of diagnosis, and the therapeutic effect is limited. Therefore, this study aimed to verify whether deoxythymidylate kinase (DTYMK) increased in HCC and was an effective therapeutic target in HCC. The findings revealed that the DTYMK level significantly increased and correlated with poor prognosis in HCC. However, nothing else is known, except that DTYMK could catalyze the phosphorylation of deoxythymidine monophosphate (dTMP) to form deoxythymidine diphosphate (dTDP). A number of experiments were performed to study the function of DTYMK in vitro and in vivo to resolve this knowledge gap. The knockdown of DTYMK was found to significantly inhibit the growth of HCC and increase the sensitivity to oxaliplatin, which is commonly used in HCC treatment. Moreover, DTYMK was found to competitively combine with miR-378a-3p to maintain the expression of MAPK activated protein kinase 2 (MAPKAPK2) and thus activate the phospho-heat shock protein 27 (phospho-HSP27)/nuclear factor NF-kappaB (NF-κB) axis, which mediated the drug resistance, proliferation of tumor cells, and infiltration of tumor-associated macrophages by inducing the expression of C-C motif chemokine ligand 5 (CCL5). Thus, this study demonstrated a new mechanism and provided a new insight into the role of mRNA in not only encoding proteins to regulate the process of life but also regulating the expression of other genes and tumor microenvironment through the competing endogenous RNA (ceRNA) mechanism.
Collapse
Affiliation(s)
- Fengze Sun
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuanyuan Liu
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tingting Gong
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Qiuzhong Pan
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tong Xiang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jingjing Zhao
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Tang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yulong Han
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mengjia Song
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yue Huang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Han Li
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuanyuan Chen
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chaopin Yang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jieying Yang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qijing Wang
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yongqiang Li
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia He
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Desheng Weng
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ruiqing Peng
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Jianchuan Xia
- Department of Biotherapy, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
Ghaznavi H, Shirvaliloo M, Zarebkohan A, Shams Z, Radnia F, Bahmanpour Z, Sargazi S, Saravani R, Shirvalilou S, Shahraki O, Shahraki S, Nazarlou Z, Sheervalilou R. An Updated Review on Implications of Autophagy and Apoptosis in Tumorigenesis: Possible Alterations in Autophagy through Engineered Nanomaterials and Their Importance in Cancer Therapy. Mol Pharmacol 2021; 100:119-143. [PMID: 33990406 DOI: 10.1124/molpharm.121.000234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Milad Shirvaliloo
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Amir Zarebkohan
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zinat Shams
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Fatemeh Radnia
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zahra Bahmanpour
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Saman Sargazi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ramin Saravani
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sakine Shirvalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sheida Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ziba Nazarlou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| |
Collapse
|
26
|
Kocak M, Ezazi Erdi S, Jorba G, Maestro I, Farrés J, Kirkin V, Martinez A, Pless O. Targeting autophagy in disease: established and new strategies. Autophagy 2021; 18:473-495. [PMID: 34241570 PMCID: PMC9037468 DOI: 10.1080/15548627.2021.1936359] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway responsible for clearing cytosolic aggregated proteins, damaged organelles or invading microorganisms. Dysfunctional autophagy leads to pathological accumulation of the cargo, which has been linked to a range of human diseases, including neurodegenerative diseases, infectious and autoimmune diseases and various forms of cancer. Cumulative work in animal models, application of genetic tools and pharmacologically active compounds, has suggested the potential therapeutic value of autophagy modulation in disease, as diverse as Huntington, Salmonella infection, or pancreatic cancer. Autophagy activation versus inhibition strategies are being explored, while the role of autophagy in pathophysiology is being studied in parallel. However, the progress of preclinical and clinical development of autophagy modulators has been greatly hampered by the paucity of selective pharmacological agents and biomarkers to dissect their precise impact on various forms of autophagy and cellular responses. Here, we summarize established and new strategies in autophagy-related drug discovery and indicate a path toward establishing a more efficient discovery of autophagy-selective pharmacological agents. With this knowledge at hand, modern concepts for therapeutic exploitation of autophagy might become more plausible. Abbreviations: ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related gene; AUTAC: autophagy-targeting chimera; CNS: central nervous system; CQ: chloroquine; GABARAP: gamma-aminobutyric acid type A receptor-associated protein; HCQ: hydroxychloroquine; LYTAC: lysosome targeting chimera; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NDD: neurodegenerative disease; PDAC: pancreatic ductal adenocarcinoma; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; PROTAC: proteolysis-targeting chimera; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Muhammed Kocak
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | | | | | - Inés Maestro
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain
| | | | - Vladimir Kirkin
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | - Ana Martinez
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Neurodegenerativas (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Ole Pless
- Fraunhofer ITMP ScreeningPort, Hamburg, Germany
| |
Collapse
|
27
|
Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies. Molecules 2021; 26:molecules26113157. [PMID: 34070502 PMCID: PMC8198360 DOI: 10.3390/molecules26113157] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/15/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) is a specific transcription factor with potent effects on the regulation of antioxidant gene expression that modulates cell hemostasis under various conditions in tissues. However, the effects of Nrf2 on gastric cancer (GC) are not fully elucidated and understood. Evidence suggests that uncontrolled Nrf2 expression and activation has been observed more frequently in malignant tumors, including GC cells, which is then associated with increased antioxidant capacity, chemoresistance, and poor clinical prognosis. Moreover, Nrf2 inhibitors and the associated modulation of tumor cell redox balance have shown that Nrf2 also has beneficial effects on the therapy of various cancers, including GC. Based on previous findings on the important role of Nrf2 in GC therapy, it is of great interest to scientists in basic and clinical tumor research that Nrf2 can be active as both an oncogene and a tumor suppressor depending on different background situations.
Collapse
|
28
|
Autophagy, an accomplice or antagonist of drug resistance in HCC? Cell Death Dis 2021; 12:266. [PMID: 33712559 PMCID: PMC7954824 DOI: 10.1038/s41419-021-03553-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a highly lethal malignancy characterized by poor prognosis and a low 5-year survival rate. Drug treatment is proving to be effective in anti-HCC. However, only a small number of HCC patients exhibit sensitive responses, and drug resistance occurs frequently in advanced patients. Autophagy, an evolutionary process responsible for the degradation of cellular substances, is closely associated with the acquisition and maintenance of drug resistance for HCC. This review focuses on autophagic proteins and explores the intricate relationship between autophagy and cancer stem cells, tumor-derived exosomes, and noncoding RNA. Clinical trials involved in autophagy inhibition combined with anticancer drugs are also concerned.
Collapse
|
29
|
Giulitti F, Petrungaro S, Mandatori S, Tomaipitinca L, de Franchis V, D'Amore A, Filippini A, Gaudio E, Ziparo E, Giampietri C. Anti-tumor Effect of Oleic Acid in Hepatocellular Carcinoma Cell Lines via Autophagy Reduction. Front Cell Dev Biol 2021; 9:629182. [PMID: 33614661 PMCID: PMC7892977 DOI: 10.3389/fcell.2021.629182] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Oleic acid (OA) is a component of the olive oil. Beneficial health effects of olive oil are well-known, such as protection against liver steatosis and against some cancer types. In the present study, we focused on OA effects in hepatocellular carcinoma (HCC), investigating responses to OA treatment (50–300 μM) in HCC cell lines (Hep3B and Huh7.5) and in a healthy liver-derived human cell line (THLE-2). Upon OA administration higher lipid accumulation, perilipin-2 increase, and autophagy reduction were observed in HCC cells as compared to healthy cells. OA in the presence of 10% FBS significantly reduced viability of HCC cell lines at 300 μM through Alamar Blue staining evaluation, and reduced cyclin D1 expression in a dose-dependent manner while it was ineffective on healthy hepatocytes. Furthermore, OA increased cell death by about 30%, inducing apoptosis and necrosis in HCC cells but not in healthy hepatocytes at 300 μM dosage. Moreover, OA induced senescence in Hep3B, reduced P-ERK in both HCC cell lines and significantly inhibited the antiapoptotic proteins c-Flip and Bcl-2 in HCC cells but not in healthy hepatocytes. All these results led us to conclude that different cell death processes occur in these two HCC cell lines upon OA treatment. Furthermore, 300 μM OA significantly reduced the migration and invasion of both HCC cell lines, while it has no effects on healthy cells. Finally, we investigated autophagy role in OA-dependent effects by using the autophagy inducer torin-1. Combined OA/torin-1 treatment reduced lipid accumulation and cell death as compared to single OA treatment. We therefore concluded that OA effects in HCC cells lines are, at least, in part dependent on OA-induced autophagy reduction. In conclusion, we report for the first time an autophagy dependent relevant anti-cancer effect of OA in human hepatocellular carcinoma cell lines.
Collapse
Affiliation(s)
- Federico Giulitti
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Simonetta Petrungaro
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Sara Mandatori
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Luana Tomaipitinca
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Valerio de Franchis
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella D'Amore
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonio Filippini
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Elio Ziparo
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Claudia Giampietri
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
30
|
Kouroumalis E, Voumvouraki A, Augoustaki A, Samonakis DN. Autophagy in liver diseases. World J Hepatol 2021; 13:6-65. [PMID: 33584986 PMCID: PMC7856864 DOI: 10.4254/wjh.v13.i1.6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is the liver cell energy recycling system regulating a variety of homeostatic mechanisms. Damaged organelles, lipids and proteins are degraded in the lysosomes and their elements are re-used by the cell. Investigations on autophagy have led to the award of two Nobel Prizes and a health of important reports. In this review we describe the fundamental functions of autophagy in the liver including new data on the regulation of autophagy. Moreover we emphasize the fact that autophagy acts like a two edge sword in many occasions with the most prominent paradigm being its involvement in the initiation and progress of hepatocellular carcinoma. We also focused to the implication of autophagy and its specialized forms of lipophagy and mitophagy in the pathogenesis of various liver diseases. We analyzed autophagy not only in well studied diseases, like alcoholic and nonalcoholic fatty liver and liver fibrosis but also in viral hepatitis, biliary diseases, autoimmune hepatitis and rare diseases including inherited metabolic diseases and also acetaminophene hepatotoxicity. We also stressed the different consequences that activation or impairment of autophagy may have in hepatocytes as opposed to Kupffer cells, sinusoidal endothelial cells or hepatic stellate cells. Finally, we analyzed the limited clinical data compared to the extensive experimental evidence and the possible future therapeutic interventions based on autophagy manipulation.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71110, Greece
| | - Argryro Voumvouraki
- 1 Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54636, Greece
| | - Aikaterini Augoustaki
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece
| | - Dimitrios N Samonakis
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece.
| |
Collapse
|
31
|
Tang Y, Chen K, Luan X, Zhang J, Liu R, Zheng X, Xie S, Ke H, Zhang X, Chen W. Knockdown of eukaryotic translation initiation factor 5A2 enhances the therapeutic efficiency of doxorubicin in hepatocellular carcinoma cells by triggering lethal autophagy. Int J Oncol 2020; 57:1368-1380. [PMID: 33174013 PMCID: PMC7646588 DOI: 10.3892/ijo.2020.5143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an invasive malignant neoplasm with a poor prognosis. The development of chemoresistance severely obstructs the chemotherapeutic efficiency of HCC treatment. Therefore, understanding the mechanisms of chemoresistance is important for improving the outcomes of patients with HCC. Eukaryotic translation initiation factor 5A2 (eIF5A2), which is considered to be an oncogene, has been reported to mediate chemoresistance in various types of cancer; however, its precise role in HCC remains unclear. Accumulating evidence has suggested that autophagy serves a dual role in cancer chemotherapy. The present study aimed to investigate the role of autophagy in eIF5A2‑mediated doxorubicin resistance in HCC. High expression levels of eIF5A2 in human HCC tissues were observed by immunohistochemistry using a tissue microarray, which was consistent with the results of reverse transcription‑quantitative PCR analysis in paired HCC and adjacent healthy tissues. HCC patient‑derived tumor xenograft mouse model was used for the in vivo study, and knockdown of eIF5A2 effectively enhanced the efficacy of doxorubicin chemotherapy compared with that in the control group. Notably, eIF5A2 served as a repressor in regulating autophagy under chemotherapy. Silencing of eIF5A2 induced doxorubicin sensitivity in HCC cells by triggering lethal autophagy. In addition, 5‑ethynyl‑2'‑deoxyuridine, lactate dehydrogenase release assay and calcein‑AM/PI staining were used to determine the enhanced autophagic cell death induced by the silencing of eIF5A2 under doxorubicin treatment. Suppression of autophagy attenuated the sensitivity of HCC cells to doxorubicin induced by eIF5A2 silencing. The results also demonstrated that knockdown of the Beclin 1 gene, which is an autophagy regulator, reversed the enhanced autophagic cell death and doxorubicin sensitivity induced by eIF5A2 silencing. Taken together, these results suggested eIF5A2 may mediate the chemoresistance of HCC cells by suppressing autophagic cell death under chemotherapy through a Beclin 1‑dependent pathway, and that eIF5A2 may be a novel potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Yuexiao Tang
- Department of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| | - Ke Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016
| | - Xiaorui Luan
- Department of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| | - Jinyan Zhang
- Department of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| | - Rongrong Liu
- Division of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003
| | - Xiaoxiao Zheng
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| | - Shangzhi Xie
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| | - Haiping Ke
- Department of Biology, Ningbo College of Health Sciences, Ningbo, Zhejiang 315100, P.R. China
| | - Xianning Zhang
- Department of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012
| |
Collapse
|
32
|
Triangular Relationship between p53, Autophagy, and Chemotherapy Resistance. Int J Mol Sci 2020; 21:ijms21238991. [PMID: 33256191 PMCID: PMC7730978 DOI: 10.3390/ijms21238991] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy and radiation often induce a number of cellular responses, such as apoptosis, autophagy, and senescence. One of the major regulators of these processes is p53, an essential tumor suppressor that is often mutated or lost in many cancer types and implicated in early tumorigenesis. Gain of function (GOF) p53 mutations have been implicated in increased susceptibility to drug resistance, by compromising wildtype anti-tumor functions of p53 or modulating key p53 processes that confer chemotherapy resistance, such as autophagy. Autophagy, a cellular survival mechanism, is initially induced in response to chemotherapy and radiotherapy, and its cytoprotective nature became the spearhead of a number of clinical trials aimed to sensitize patients to chemotherapy. However, increased pre-clinical studies have exemplified the multifunctional role of autophagy. Additionally, compartmental localization of p53 can modulate induction or inhibition of autophagy and may play a role in autophagic function. The duality in p53 function and its effects on autophagic function are generally not considered in clinical trial design or clinical therapeutics; however, ample pre-clinical studies suggest they play a role in tumor responses to therapy and drug resistance. Further inquiry into the interconnection between autophagy and p53, and its effects on chemotherapeutic responses may provide beneficial insights on multidrug resistance and novel treatment regimens for chemosensitization.
Collapse
|
33
|
Gąsiorkiewicz BM, Koczurkiewicz-Adamczyk P, Piska K, Pękala E. Autophagy modulating agents as chemosensitizers for cisplatin therapy in cancer. Invest New Drugs 2020; 39:538-563. [PMID: 33159673 PMCID: PMC7960624 DOI: 10.1007/s10637-020-01032-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023]
Abstract
Although cisplatin is one of the most common antineoplastic drug, its successful utilisation in cancer treatment is limited by the drug resistance. Multiple attempts have been made to find potential cisplatin chemosensitisers which would overcome cancer cells resistance thus improving antineoplastic efficacy. Autophagy modulation has become an important area of interest regarding the aforementioned topic. Autophagy is a highly conservative cellular self-digestive process implicated in response to multiple environmental stressors. The high basal level of autophagy is a common phenomenon in cisplatin-resistant cancer cells which is thought to grant survival benefit. However current evidence supports the role of autophagy in either promoting or limiting carcinogenesis depending on the context. This encourages the search of substances modulating the process to alleviate cisplatin resistance. Such a strategy encompasses not only simple autophagy inhibition but also harnessing the process to induce autophagy-dependent cell death. In this paper, we briefly describe the mechanism of cisplatin resistance with a special emphasis on autophagy and we give an extensive literature review of potential substances with cisplatin chemosensitising properties related to autophagy modulation.
Collapse
Affiliation(s)
- Bartosz Mateusz Gąsiorkiewicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Paulina Koczurkiewicz-Adamczyk
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Kamil Piska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| |
Collapse
|
34
|
Morin Hydrate Sensitizes Hepatoma Cells and Xenograft Tumor towards Cisplatin by Downregulating PARP-1-HMGB1 Mediated Autophagy. Int J Mol Sci 2020; 21:ijms21218253. [PMID: 33158052 PMCID: PMC7885522 DOI: 10.3390/ijms21218253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
The cross-talk between apoptosis and autophagy influences anticancer drug sensitivity and cellular death in various cancer cell lines. However, the fundamental mechanisms behind this phenomenon are still unidentified. We demonstrated anti-cancerous role of cisplatin (CP) and morin hydrate (Mh) as an individual and/or in combination (CP-Mh) in hepatoma cells and tumor model. Exposure of CP resulted in the production of intracellular reactive oxygen species (ROS)-mediated cellular vacuolization, expansion of mitochondria membrane and activation of endoplasmic reticulum (ER)-stress. Consequently, Cyt c translocation led to the increase of Bax/Bcl-2 ratio, which simultaneously triggered caspase-mediated cellular apoptosis. In addition, CP-induced PARP-1 activation led to ADP-ribosylation of HMGB1, which consequently developed autophagy as evident by the LC3I/II ratio. Chemically-induced inhibition of autophagy marked by increased cell death signified a protective role of autophagy against CP treatment. CP-Mh abrogates the PARP-1 expression and significantly reduced HMGB1-cytoplasmic translocation with subsequent inhibition of the HMGB1-Beclin1 complex formation. In the absence of PARP-1, a reduced HMGB1 mediated autophagy was observed followed by induced caspase-dependent apoptosis. To confirm the role of PARP-1-HMGB1 signaling in autophagy, we used the PARP-1 inhibitor, 4-amino-1,8-naphthalimide (ANI), HMGB1 inhibitor, ethyl pyruvate (EP), autophagy inhibitors, 3-methyl adenine (3-MA) and bafilomycin (baf) and small interfering RNAs (siRNA) to target Atg5 in combination of CP and Mh. Exposure to these inhibitors enhanced the sensitivity of HepG2 cells to CP. Collectively, our findings indicate that CP-Mh in combination served as a prominent regulator of autophagy and significant inducer of apoptosis that maintains a homeostatic balance towards HepG2 cells and the subcutaneous tumor model.
Collapse
|
35
|
Fan J, Shi Y, Peng Y. Autophagy and Liver Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:497-528. [PMID: 32671772 DOI: 10.1007/978-981-15-4272-5_37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autophagy plays an important role in the physiology and pathology of the liver. It is involved in the development of many liver diseases such as α-1-antitrypsin deficiency, chronic hepatitis virus infection, alcoholic liver disease, nonalcoholic fatty liver disease, and liver cancer. Autophagy has thus become a new target for the treatment of liver diseases. How to treat liver diseases by regulating autophagy has been a hot topic.
Collapse
Affiliation(s)
- Jia Fan
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China.
| | - Yinghong Shi
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China
| | - Yuanfei Peng
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China
| |
Collapse
|
36
|
Dinić J, Efferth T, García-Sosa AT, Grahovac J, Padrón JM, Pajeva I, Rizzolio F, Saponara S, Spengler G, Tsakovska I. Repurposing old drugs to fight multidrug resistant cancers. Drug Resist Updat 2020; 52:100713. [PMID: 32615525 DOI: 10.1016/j.drup.2020.100713] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/08/2023]
Abstract
Overcoming multidrug resistance represents a major challenge for cancer treatment. In the search for new chemotherapeutics to treat malignant diseases, drug repurposing gained a tremendous interest during the past years. Repositioning candidates have often emerged through several stages of clinical drug development, and may even be marketed, thus attracting the attention and interest of pharmaceutical companies as well as regulatory agencies. Typically, drug repositioning has been serendipitous, using undesired side effects of small molecule drugs to exploit new disease indications. As bioinformatics gain increasing popularity as an integral component of drug discovery, more rational approaches are needed. Herein, we show some practical examples of in silico approaches such as pharmacophore modelling, as well as pharmacophore- and docking-based virtual screening for a fast and cost-effective repurposing of small molecule drugs against multidrug resistant cancers. We provide a timely and comprehensive overview of compounds with considerable potential to be repositioned for cancer therapeutics. These drugs are from diverse chemotherapeutic classes. We emphasize the scope and limitations of anthelmintics, antibiotics, antifungals, antivirals, antimalarials, antihypertensives, psychopharmaceuticals and antidiabetics that have shown extensive immunomodulatory, antiproliferative, pro-apoptotic, and antimetastatic potential. These drugs, either used alone or in combination with existing anticancer chemotherapeutics, represent strong candidates to prevent or overcome drug resistance. We particularly focus on outcomes and future perspectives of drug repositioning for the treatment of multidrug resistant tumors and discuss current possibilities and limitations of preclinical and clinical investigations.
Collapse
Affiliation(s)
- Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | | | - Jelena Grahovac
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, E-38071 La Laguna, Spain.
| | - Ilza Pajeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 301724 Venezia-Mestre, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Dóm tér 10, Hungary
| | - Ivanka Tsakovska
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| |
Collapse
|
37
|
Wang X, Li M, Ren K, Xia C, Li J, Yu Q, Qiu Y, Lu Z, Long Y, Zhang Z, He Q. On-Demand Autophagy Cascade Amplification Nanoparticles Precisely Enhanced Oxaliplatin-Induced Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002160. [PMID: 32596861 DOI: 10.1002/adma.202002160] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/21/2020] [Indexed: 06/11/2023]
Abstract
Chemoimmunotherapy-induced antitumor immune response is highly dependent on tumor autophagy. When tumor cells are treated with chemoimmunotherapy, timely overactivated autophagy can not only lead more tumor cells to death, but also participate in the endogenous antigen presentation and immune stimulators secretion of dying cells, thus plays a vital role. However, timely and accurately overactivated tumor autophagy during chemoimmunotherapy is of great difficulty. Here, an on-demand autophagy cascade amplification nanoparticle (ASN) is reported to boost oxaliplatin-induced cancer immunotherapy. ASN is prepared by self-assemble of autophagy-responsible C-TFG micelle and is followed by electrostatic binding of oxaliplatin prodrug (HA-OXA). After entering tumor cells, the HA-OXA shell of ASN first responds to the reduction microenvironment and releases oxaliplatin to trigger tumor immunogenic cell death and mildly stimulates tumor autophagy. Then, the exposed C-TFG micelle can sensitively respond to oxaliplatin-induced autophagy and release a powerful autophagy inducer STF-62247, which precisely transforms autophagy to "overactivated" condition, leading tumor cells to autophagic death and enhance subsequent tumor antigen processing of the dying cells. In CT26 tumor-bearing mice, ASN exhibits optimal immune stimulation and antitumor efficiency due to its on-demand autophagy induction ability.
Collapse
Affiliation(s)
- Xuhui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Kebai Ren
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Chunyu Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Jianping Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Qianwen Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Yue Qiu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Zhengze Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Yang Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
38
|
Antitumor Activity of Pt(II), Ru(III) and Cu(II) Complexes. Molecules 2020; 25:molecules25153492. [PMID: 32751963 PMCID: PMC7435640 DOI: 10.3390/molecules25153492] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/25/2022] Open
Abstract
Metal complexes are currently potential therapeutic compounds. The acquisition of resistance by cancer cells or the effective elimination of cancer-affected cells necessitates a constant search for chemical compounds with specific biological activities. One alternative option is the transition metal complexes having potential as antitumor agents. Here, we present the current knowledge about the application of transition metal complexes bearing nickel(II), cobalt(II), copper(II), ruthenium(III), and ruthenium(IV). The cytotoxic properties of the above complexes causing apoptosis, autophagy, DNA damage, and cell cycle inhibition are described in this review.
Collapse
|
39
|
Wan ZH, Jiang TY, Shi YY, Pan YF, Lin YK, Ma YH, Yang C, Feng XF, Huang LF, Kong XN, Ding ZW, Tan YX, Dong LW, Wang HY. RPB5-Mediating Protein Promotes Cholangiocarcinoma Tumorigenesis and Drug Resistance by Competing With NRF2 for KEAP1 Binding. Hepatology 2020; 71:2005-2022. [PMID: 31541481 DOI: 10.1002/hep.30962] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Cancer cell survival depends on the balance between reactive oxygen species production and scavenging, which is regulated primarily by NRF2 during tumorigenesis. Here, we demonstrate that deletion of RBP5-mediating protein (RMP) in an autonomous mouse model of intrahepatic cholangiocarcinoma (ICC) delays tumor progression. APPROACH AND RESULTS RMP-overexpressing tumor cells exhibited enhanced tolerance to oxidative stress and apoptosis. Mechanistically, RMP competes with NRF2 for binding to the Kelch domain of KEAP1 (Kelch-like ECH-associated protein 1) through the E**E motif, leading to decreased NRF2 degradation via ubiquitination, thus increasing NRF2 nuclear translocation and downstream transactivation of antioxidant genes. This RMP-KEAP1-NRF2 axis promotes ICC tumorigenesis, metastasis, and drug resistance. Consistent with these findings, the RMP level in human ICC is positively correlated with the protein level of NRF2 and is associated with poor prognosis. CONCLUSION These findings reveal that RMP is involved in the oxidative stress defense program and could be exploited for targeted cancer therapies.
Collapse
Affiliation(s)
- Zheng-Hua Wan
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China.,No. 971 Hospital of Peoples' Liberation Army Navy, Qing Dao, China
| | - Tian-Yi Jiang
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China
| | - Yuan-Yuan Shi
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China
| | - Yu-Fei Pan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China
| | - Yun-Kai Lin
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China
| | - Yun-Han Ma
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China
| | - Chun Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China.,Children's Hospital of Soochow University, Suzhou, China
| | - Xiao-Fan Feng
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China
| | - Li-Feng Huang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Wen Ding
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China
| | - Ye-Xiong Tan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China
| | - Li-Wei Dong
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China
| | - Hong-Yang Wang
- National Center for Liver Cancer, the Second Military Medical University, Shanghai, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, The Second Military Medical University & Ministry of Education, Shanghai, China
| |
Collapse
|
40
|
Modulation of the Autophagy-lysosomal Pathway in Hepatocellular Carcinoma Using Small Molecules. Molecules 2020; 25:molecules25071580. [PMID: 32235537 PMCID: PMC7181071 DOI: 10.3390/molecules25071580] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/28/2020] [Accepted: 03/29/2020] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for approximately 90% of all cases of primary liver cancer; it is the third most frequent cause of cancer-related death worldwide. In early-stage disease, surgical resection and liver transplantation are considered curative treatments. However, the majority of HCC patients present with advanced-stage disease that is treated using palliative systemic therapy. Since HCC is heterogeneous owing to its multiple etiologies, various risk factors, and inherent resistance to chemotherapy, the development of an effective systemic treatment strategy for HCC remains a considerable challenge. Autophagy is a lysosome-dependent catabolic degradation pathway that is essential for maintaining cellular energy homeostasis. Autophagy dysfunction is closely linked with the pathogenesis of various cancers; therefore, the discovery of small molecules that can modulate autophagy has attracted considerable interest in the development of a systemic treatment strategy for advanced HCC. Here, we reviewed the roles of autophagy in HCC and the recent advances regarding small molecules that target autophagy regulatory mechanisms.
Collapse
|
41
|
Kiruthiga C, Devi KP, Nabavi SM, Bishayee A. Autophagy: A Potential Therapeutic Target of Polyphenols in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12030562. [PMID: 32121322 PMCID: PMC7139730 DOI: 10.3390/cancers12030562] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved biological phenomenon that maintains cellular homeostasis through the clearing of damaged cellular components under cellular stress and offers the cell building blocks for cellular survival. Aberrations in autophagy subsidize to various human pathologies, such as dementia, cardiovascular diseases, leishmaniosis, influenza, hepatic diseases, and cancer, including hepatocellular carcinoma (HCC). HCC is the fifth common mortal type of liver cancer globally, with an inhomogeneous topographical distribution and highest incidence tripled in men than women. Existing treatment procedures with liver cancer patients result in variable success rates and poor prognosis due to their drug resistance and toxicity. One of the pathophysiological mechanisms that are targeted during the development of anti-liver cancer drugs is autophagy. Generally, overactivated autophagy may lead to a non-apoptotic form of programmed cell death (PCD) or autophagic cell death or type II PCD. Emerging evidence suggests that manipulation of autophagy could induce type II PCD in cancer cells, acting as a potential tumor suppressor. Hence, altering autophagic signaling offers new hope for the development of novel drugs for the therapy of resistant cancer cells. Natural polyphenolic compounds, including flavonoids and non-flavonoids, execute their anticarcinogenic mechanism through upregulating tumor suppressors and autophagy by modulating canonical (Beclin-1-dependent) and non-canonical (Beclin-1-independent) signaling pathways. Additionally, there is evidence signifying that plant polyphenols target angiogenesis and metastasis in HCC via interference with multiple intracellular signals and decrease the risk against HCC. The current review offers a comprehensive understanding of how natural polyphenolic compounds exhibit their anti-HCC effects through regulation of autophagy, the non-apoptotic mode of cell death.
Collapse
Affiliation(s)
- Chandramohan Kiruthiga
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| | - Seyed M. Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| |
Collapse
|
42
|
Zeng Y, Guo Z, Hu Z, Liu M, Chen Y, Chen S, Peng B, Zhang P, Wu Z, Luo H, Zhong F, Jiang K, Lu Y, Yuan G, He S. FGD1 exhibits oncogenic properties in hepatocellular carcinoma through regulating cell morphology, autophagy and mitochondrial function. Biomed Pharmacother 2020; 125:110029. [PMID: 32106378 DOI: 10.1016/j.biopha.2020.110029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
Faciogenital Dysplasia 1 (FGD1) has been involved in a variety of biological processes, including cytoskeleton restructuring, cell morphology, cell cycle progression, and cell polarity. Abnormal expression of FGD1 was also identified in several types of cancers, indicating its critical role in the development of cancers. However, little is known about the role of FGD1 in hepatocellular carcinoma (HCC). In this study, the expression of FGD1 in HCC was mined with the RNA sequencing data from the cancer genome atlas. By over-expressing or knocking down of FGD1, the effects of FGD1 on the malignant behavior of HCC were evaluated both in vitro and in vivo. We find that FGD1 is up-regulated in HCC and correlated with the development and prognosis of HCC. By over-expressing or knocking down of FGD1, the effects of FGD1 on the malignant behavior of HCC were evaluated both in vitro and in vivo. Knockdown of FGD1 remarkably inhibits the malignant behaviors and causes morphological disorder of pseudopodia, autophagy inhibition and mitochondrial dyfunction in HCC cells. Further investigation shows that Cdc42, a Rho GTPase, plays a role in these processes. Overexpression of FGD1 significantly promotes the oncogenic properties of HCC cells. Collectively, these findings reveal that FGD1 exhibits oncogenic properties in HCC through regulating cell morphology, autophagy and mitochondrial function, suggesting that FGD1 may serve as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yonglian Zeng
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China; Basic Medical School of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhenya Guo
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhigao Hu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Mingjiang Liu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yubing Chen
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shilian Chen
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bo Peng
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Peng Zhang
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhan Wu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hongliang Luo
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Fudi Zhong
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Keqing Jiang
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology, 1088 Xili Xueyuan Avenue, Shenzhen, 518055, Guangdong, China.
| | - Guandou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
43
|
Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells. Mol Med 2019; 25:49. [PMID: 31726966 PMCID: PMC6854621 DOI: 10.1186/s10020-019-0116-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022] Open
Abstract
Background Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known to promote cell survival, including autophagy. As such, targeting these survival pathways may represent new vulnerabilities of GBM after treatment with TMZ. Methods Using the T98G human glioma cell line, we assessed the molecular signaling associated with TMZ treatment, the cellular consequences of using the pan-PI3K inhibitor PX-866, and performed clonogenic assays to determine the effect sequential treatment of TMZ and PX-866 had on colony formation. Additionally, we also use subcutaneous GBM patient derived xenograft (PDX) tumors to show relative LC3 protein expression and correlations between survival pathways and molecular markers which dictate clinical responsiveness to TMZ. Results Here, we report that TMZ can induce autophagic flux in T98G glioma cells. GBM patient-derived xenograft (PDX) tumors treated with TMZ also display an increase in the autophagosome marker LC3 II. Additionally, O6-methylguanine-DNA-methyltransferase (MGMT) expression correlates with PI3K/AKT activity, suggesting that patients with inherent resistance to TMZ (MGMT-high) would benefit from PI3K/AKT inhibitors in addition to TMZ. Accordingly, we have identified that the blood-brain barrier (BBB) penetrant pan-PI3K inhibitor, PX-866, is an early-stage inhibitor of autophagic flux, while maintaining its ability to inhibit PI3K/AKT signaling in glioma cells. Lastly, due to the induction of autophagic flux by TMZ, we provide evidence for sequential treatment of TMZ followed by PX-866, rather than combined co-treatment, as a means to shut down autophagy-induced survival in GBM cells and to enhance apoptosis. Conclusions The understanding of how TMZ induces survival pathways, such as autophagy, may offer new therapeutic vulnerabilities and opportunities to use sequential inhibition of alternate pro-survival pathways that regulate autophagy. As such, identification of additional ways to inhibit TMZ-induced autophagy could enhance the efficacy of TMZ.
Collapse
|
44
|
Ho CJ, Gorski SM. Molecular Mechanisms Underlying Autophagy-Mediated Treatment Resistance in Cancer. Cancers (Basel) 2019; 11:E1775. [PMID: 31717997 PMCID: PMC6896088 DOI: 10.3390/cancers11111775] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Despite advances in diagnostic tools and therapeutic options, treatment resistance remains a challenge for many cancer patients. Recent studies have found evidence that autophagy, a cellular pathway that delivers cytoplasmic components to lysosomes for degradation and recycling, contributes to treatment resistance in different cancer types. A role for autophagy in resistance to chemotherapies and targeted therapies has been described based largely on associations with various signaling pathways, including MAPK and PI3K/AKT signaling. However, our current understanding of the molecular mechanisms underlying the role of autophagy in facilitating treatment resistance remains limited. Here we provide a comprehensive summary of the evidence linking autophagy to major signaling pathways in the context of treatment resistance and tumor progression, and then highlight recently emerged molecular mechanisms underlying autophagy and the p62/KEAP1/NRF2 and FOXO3A/PUMA axes in chemoresistance.
Collapse
Affiliation(s)
- Cally J. Ho
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada;
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada;
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
45
|
Redox-Mediated Mechanism of Chemoresistance in Cancer Cells. Antioxidants (Basel) 2019; 8:antiox8100471. [PMID: 31658599 PMCID: PMC6826977 DOI: 10.3390/antiox8100471] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular reactive oxygen species (ROS) status is stabilized by a balance of ROS generation and elimination called redox homeostasis. ROS is increased by activation of endoplasmic reticulum stress, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family members and adenosine triphosphate (ATP) synthesis of mitochondria. Increased ROS is detoxified by superoxide dismutase, catalase, and peroxiredoxins. ROS has a role as a secondary messenger in signal transduction. Cancer cells induce fluctuations of redox homeostasis by variation of ROS regulated machinery, leading to increased tumorigenesis and chemoresistance. Redox-mediated mechanisms of chemoresistance include endoplasmic reticulum stress-mediated autophagy, increased cell cycle progression, and increased conversion to metastasis or cancer stem-like cells. This review discusses changes of the redox state in tumorigenesis and redox-mediated mechanisms involved in tolerance to chemotherapeutic drugs in cancer.
Collapse
|
46
|
miR-199a-5p Represses Protective Autophagy and Overcomes Chemoresistance by Directly Targeting DRAM1 in Acute Myeloid Leukemia. JOURNAL OF ONCOLOGY 2019; 2019:5613417. [PMID: 31636666 PMCID: PMC6766143 DOI: 10.1155/2019/5613417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 08/19/2019] [Indexed: 12/26/2022]
Abstract
Chemotherapy resistance is still a primary clinical obstacle to the successful treatment of acute myeloid leukemia (AML). The underlying mechanisms of drug resistance are complicated and have not been fully understood. Here, we found that miR-199a-5p levels were significantly reduced in refractory/relapsed AML patients compared to those who achieved complete remission after chemotherapy. Consistently, miR-199a-5p was markedly decreased in Adriamycin-resistant AML K562/ADM cells in contrast with Adriamycin-sensitive K562 cells, and its decrement dramatically correlated with the chemoresistance of AML cells. Furthermore, we demonstrated that the basic and Adriamycin-induced autophagic activity in K562/ADM cells was higher than that in K562 cells. This inducible autophagy played a prosurvival role and contributed to the development of acquired drug resistance. Importantly, we investigated that miR-199a-5p could negatively regulate autophagy, at least in part, by inhibiting damage regulator autophagy modulator (DRAM1) expression at both the transcriptional and posttranscriptional level. miR-199a-5p bound directly to the 3'-UTR of DRAM1 mRNA which was a functional target of miR-199a-5p. Indeed, downregulation of DRAM1 gene by siRNA in K562/ADM cells resulted in autophagy suppression and chemosensitivity restoration. These results revealed that the miR-199a-5p/DRAM1/autophagy signaling represented a novel pathway regulating chemoresistance, indicating a potential therapeutic strategy for the intervention in drug-resistant AML.
Collapse
|
47
|
Oxidative Stress-Driven Autophagy acROSs Onset and Therapeutic Outcome in Hepatocellular Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6050123. [PMID: 31205585 PMCID: PMC6530208 DOI: 10.1155/2019/6050123] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/28/2019] [Indexed: 12/22/2022]
Abstract
Reactive oxygen species- (ROS-) mediated autophagy physiologically contributes to management of cell homeostasis in response to mild oxidative stress. Cancer cells typically engage autophagy downstream of ROS signaling derived from hypoxia and starvation, which are harsh environmental conditions that need to be faced for cancer development and progression. Hepatocellular carcinoma (HCC) is a solid tumor for which several environmental risk factors, particularly viral infections and alcohol abuse, have been shown to promote carcinogenesis via augmentation of oxidative stress. In addition, ROS burst in HCC cells frequently takes place after administration of therapeutic compounds that promote apoptotic cell death or even autophagic cell death. The interplay between ROS and autophagy (i) in the disposal of dysfunctional mitochondria via mitophagy, as a tumor suppressor mechanism, or (ii) in the cell survival adaptive response elicited by chemotherapeutic interventions, as a tumor-promoting event, will be depicted in this review in relation to HCC development and progression.
Collapse
|
48
|
Jiang J, Chen S, Li K, Zhang C, Tan Y, Deng Q, Chai Y, Wang X, Chen G, Feng K, Zhang L, Xie CM, Ma K. Targeting autophagy enhances heat stress-induced apoptosis via the ATP-AMPK-mTOR axis for hepatocellular carcinoma. Int J Hyperthermia 2019; 36:499-510. [PMID: 31007109 DOI: 10.1080/02656736.2019.1600052] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Shihan Chen
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Kun Li
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Chang Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Yunhua Tan
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Qingsong Deng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Yuelong Chai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Xiaofei Wang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Geng Chen
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Kai Feng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Leida Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Chuan-Ming Xie
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Kuansheng Ma
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| |
Collapse
|
49
|
Abstract
Resistance to therapy is one of the prime causes for treatment failure in cancer and recurrent disease. In recent years, autophagy has emerged as an important cell survival mechanism in response to different stress conditions that are associated with cancer treatment and aging. Autophagy is an evolutionary conserved catabolic process through which damaged cellular contents are degraded after uptake into autophagosomes that subsequently fuse with lysosomes for cargo degradation, thereby alleviating stress. In addition, autophagy serves to maintain cellular homeostasis by enriching nutrient pools. Although autophagy can act as a double-edged sword at the interface of cell survival and cell death, increasing evidence suggest that in the context of cancer therapy-induced stress responses, it predominantly functions as a cell survival mechanism. Here, we provide an up-to-date overview on our current knowledge of the role of pro-survival autophagy in cancer therapy at the preclinical and clinical stages and delineate the molecular mechanisms of autophagy regulation in response to therapy-related stress conditions. A better understanding of the interplay of cancer therapy and autophagy may allow to unveil new targets and avenues for an improved treatment of therapy-resistant tumors in the foreseeable future.
Collapse
|
50
|
Hounjet J, Habets R, Schaaf MB, Hendrickx TC, Barbeau LMO, Yahyanejad S, Rouschop KM, Groot AJ, Vooijs M. The anti-malarial drug chloroquine sensitizes oncogenic NOTCH1 driven human T-ALL to γ-secretase inhibition. Oncogene 2019; 38:5457-5468. [PMID: 30967635 DOI: 10.1038/s41388-019-0802-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 01/30/2019] [Accepted: 03/19/2019] [Indexed: 12/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from T-cell progenitors. Although current treatments, including chemotherapy and glucocorticoids, have significantly improved survival, T-ALL remains a fatal disease and new treatment options are needed. Since more than 60% of T-ALL cases bear oncogenic NOTCH1 mutations, small molecule inhibitors of NOTCH1 signalling; γ-secretase inhibitors (GSI), are being actively investigated for the treatment of T-ALL. Unfortunately, GSI have shown limited clinical efficacy and dose-limiting toxicities. We hypothesized that by combining known drugs, blocking NOTCH activity through another mechanism, may synergize with GSI enabling equal efficacy at a lower concentration. Here, we show that the clinically used anti-malarial drug chloroquine (CQ), an inhibitor of lysosomal function and autophagy, decreases T-ALL cell viability and proliferation. This effect of CQ was not observed in GSI-resistant T-ALL cell lines. Mechanistically, CQ impairs the redox balance, induces ds DNA breaks and activates the DNA damage response. CQ also interferes with intracellular trafficking and processing of oncogenic NOTCH1. Interestingly, we show for the first time that the addition of CQ to γ-secretase inhibition has a synergistic therapeutic effect on T-ALL and reduces the concentration of GSI required to obtain a reduction in cell viability and a block of proliferation. Overall, our results suggest that CQ may be a promising repurposed drug in the treatment of T-ALL, as a single treatment or in combination with GSI, increasing the therapeutic ratio.
Collapse
Affiliation(s)
- Judith Hounjet
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.,MAASTRO Clinic, Maastricht, The Netherlands
| | - Roger Habets
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marco B Schaaf
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Tessa C Hendrickx
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Lydie M O Barbeau
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Sanaz Yahyanejad
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Kasper M Rouschop
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Arjan J Groot
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marc Vooijs
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|