1
|
Liu HS, Wang YP, Lin PW, Chu ML, Lan SH, Wu SY, Lee YR, Chang HY. The role of Atg5 gene in tumorigenesis under autophagy deficiency conditions. Kaohsiung J Med Sci 2024; 40:631-641. [PMID: 38826147 DOI: 10.1002/kjm2.12853] [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: 01/16/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/04/2024] Open
Abstract
Autophagy is a self-recycling machinery to maintain cellular homeostasis by degrading harmful materials in the cell. Autophagy-related gene 5 (Atg5) is required for autophagosome maturation. However, the role of Atg5 in tumorigenesis under autophagy deficient conditions remains unclear. This study focused on the autophagy-independent role of Atg5 and the underlying mechanism in tumorigenesis. We demonstrated that knockout of autophagy-related genes including Atg5, Atg7, Atg9, and p62 in mouse embryonic fibroblast (MEF) cells consistently decreased cell proliferation and motility, implying that autophagy is required to maintain diverse cellular functions. An Atg7 knockout MEF (Atg7-/- MEF) cell line representing deprivation of autophagy function was used to clarify the role of Atg5 transgene in tumorigenesis. We found that Atg5-overexpressed Atg7-/-MEF (clone A) showed increased cell proliferation, colony formation, and migration under autophagy deficient conditions. Accordingly, rescuing the autophagy deficiency of clone A by overexpression of Atg7 gene shifts the role of Atg5 from pro-tumor to anti-tumor status, indicating the dual role of Atg5 in tumorigenesis. Notably, the xenograft mouse model showed that clone A of Atg5-overexpressed Atg7-/- MEF cells induced temporal tumor formation, but could not prolong further tumor growth. Finally, biomechanical analysis disclosed increased Wnt5a secretion and p-JNK expression along with decreased β-catenin expression. In summary, Atg5 functions as a tumor suppressor to protect the cell under normal conditions. In contrast, Atg5 shifts to a pro-tumor status under autophagy deprivation conditions.
Collapse
Affiliation(s)
- Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Tropical Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Teaching and Research Center, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medial University, Kaohsiung, Taiwan
| | - Yin-Ping Wang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Wen Lin
- Tropical Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Man-Ling Chu
- Tropical Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Hui Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shan-Ying Wu
- Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Ray Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hong-Yi Chang
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Cai H, Meng Z, Yu F. The involvement of ROS-regulated programmed cell death in hepatocellular carcinoma. Crit Rev Oncol Hematol 2024; 197:104361. [PMID: 38626849 DOI: 10.1016/j.critrevonc.2024.104361] [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: 11/04/2023] [Revised: 03/11/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Reactive oxidative species (ROS) is a crucial factor in the regulation of cellular biological activity and function, and aberrant levels of ROS can contribute to the development of a variety of diseases, particularly cancer. Numerous discoveries have affirmed that this process is strongly associated with "programmed cell death (PCD)," which refers to the suicide protection mechanism initiated by cells in response to external stimuli, such as apoptosis, autophagy, ferroptosis, etc. Research has demonstrated that ROS-induced PCD is crucial for the development of hepatocellular carcinoma (HCC). These activities serve a dual function in both facilitating and inhibiting cancer, suggesting the existence of a delicate balance within healthy cells that can be disrupted by the abnormal generation of reactive oxygen species (ROS), thereby influencing the eventual advancement or regression of a tumor. In this review, we summarize how ROS regulates PCD to influence the tumorigenesis and progression of HCC. Studying how ROS-induced PCD affects the progression of HCC at a molecular level can help develop better prevention and treatment methods and facilitate the design of more effective preventative and therapeutic strategies.
Collapse
Affiliation(s)
- Hanchen Cai
- The First Afliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Ziqi Meng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China; The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
3
|
The Role of Autophagy in Breast Cancer Metastasis. Biomedicines 2023; 11:biomedicines11020618. [PMID: 36831154 PMCID: PMC9953203 DOI: 10.3390/biomedicines11020618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Patient morbidity and mortality is significantly increased in metastatic breast cancer. The metastasis process of breast cancer is very complicated and is delicately controlled by various factors. Autophagy is one of the important regulatory factors affecting metastasis in breast cancer by engaging in cell mobility, metabolic adaptation, tumor dormancy, and cancer stem cells. Here, we discuss the effects of autophagy on metastasis in breast cancer and assess the potential use of autophagy modulators for metastasis treatment.
Collapse
|
4
|
Chrysophanol-Induced Autophagy Disrupts Apoptosis via the PI3K/Akt/mTOR Pathway in Oral Squamous Cell Carcinoma Cells. Medicina (B Aires) 2022; 59:medicina59010042. [PMID: 36676666 PMCID: PMC9864245 DOI: 10.3390/medicina59010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Background and Objectives: Natural products are necessary sources for drug discovery and have contributed to cancer chemotherapy over the past few decades. Furthermore, substances derived from plants have fewer side effects. Chrysophanol is an anthraquinone derivative that is isolated from rhubarb. Although the anticancer effect of chrysophanol on several cancer cells has been reported, studies on the antitumor effect of chrysophanol on oral squamous-cell carcinoma (OSCC) cells have yet to be elucidated. Therefore, in this study, we investigated the anticancer effect of chrysophanol on OSCC cells (CAL-27 and Ca9-22) via apoptosis and autophagy, among the cell death pathways. Results: It was found that chrysophanol inhibited the growth and viability of CAL-27 and Ca9-22 and induced apoptosis through the intrinsic pathway. It was also found that chrysophanol activates autophagy-related factors (ATG5, beclin-1, and P62/SQSTM1) and LC3B conversion. That is, chrysophanol activated both apoptosis and autophagy. Here, we focused on the roles of chrysophanol-induced apoptosis and the autophagy pathway. When the autophagy inhibitor 3-MA and PI3K/Akt inhibitor were used to inhibit the autophagy induced by chrysophanol, it was confirmed that the rate of apoptosis significantly increased. Therefore, we confirmed that chrysophanol induces apoptosis and autophagy at the same time, and the induced autophagy plays a role in interfering with apoptosis processes. Conclusions: Therefore, the potential of chrysophanol as an excellent anticancer agent in OSCC was confirmed via this study. Furthermore, the combined treatment of drugs that can inhibit chrysophanol-induced autophagy is expected to have a tremendous synergistic effect in overcoming oral cancer.
Collapse
|
5
|
Gui F, Yu X, Wu Y, Wu C, Zhang Y. Mechanism of LncHOTAIR Regulating Proliferation, Apoptosis, and Autophagy of Lymphoma Cells through hsa-miR-6511b-5p/ATG7 Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2166605. [PMID: 36248425 PMCID: PMC9560808 DOI: 10.1155/2022/2166605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
Objective To explore the role of LncHOTAIR in apoptosis and autophagy in lymphoma. Methods The interaction between LncHOTAIR and miR-6511b-5p, as well as between miR-6511b-5p and ATG7, was verified by a dual luciferase assay. LncHOTAIR overexpression lentivirus was transducted and siATG7s were transfected into Raji and BJAB lymphoma cells, and the efficiency was verified by qPCR. Lymphocyte proliferation was detected by the cell counting kit-8 (CCK8) test, and autophagy was detected by transmission electron microscopy. The protein expressions of ULK1, Beclin1, ATG7, LC3, Bax, cleaved-caspase 3, and Bcl-2 were detected using Western blots. Results There was a targeting relationship between LncHOTAIR and miR-6511b-5p and between miR-6511b-5p and ATG7. LncHOTAIR overexpression promoted the proliferation and autophagy of Raji and BJAB cells, significantly upregulated ATG7, Beclin1, ULK1, Bcl-2, and LC3-II/LC3-I levels, and downregulated Bax and cleaved-caspase3 levels. siATG7 significantly inhibited the proliferation and autophagy of Raji and BJAB cells and promoted their apoptosis. Conclusion LncHOTAIR/hsa-miR-6511b-5p/ATG7 could regulate the proliferation, apoptosis, and autophagy of Raji and BJAB lymphoma cells.
Collapse
Affiliation(s)
- Fu Gui
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xinyi Yu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yemeng Wu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chao Wu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yulan Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
6
|
Wang X, Shang M, Sun X, Guo L, Xiao S, Shi D, Meng D, Zhao Y, Yang L, Jiang C, Li J. Dual-responsive nanodroplets combined with ultrasound-targeted microbubble destruction suppress tumor growth and metastasis via autophagy blockade. J Control Release 2022; 343:66-77. [DOI: 10.1016/j.jconrel.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
|
7
|
Wang Y, Li Z, Xu S, Li W, Chen M, Jiang M, Fan X. LncRNA FIRRE functions as a tumor promoter by interaction with PTBP1 to stabilize BECN1 mRNA and facilitate autophagy. Cell Death Dis 2022. [PMID: 35110535 DOI: 10.1038/s41419-022-04509-1.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Long non-coding RNAs (lncRNAs) play critical functions in various cancers. Firre intergenic repeating RNA element (FIRRE), a lncRNA located in the nucleus, was overexpressed in colorectal cancer (CRC). However, the detailed mechanism of FIRRE in CRC remains elusive. Results of RNA sequence and qPCR illustrated overexpression of FIRRE in CRC cell lines and tissues. The aberrant expression of FIRRE was correlated with the migration, invasion, and proliferation in cell lines. In accordance, it was also associated with lymphatic metastasis and distant metastasis in patients with CRC. FIRRE was identified to physically interact with Polypyrimidine tract-binding protein (PTBP1) by RNA pull-down and RNA immunoprecipitation (RIP). Overexpression of FIRRE induced the translocation of PTBP1 from nucleus to cytoplasm, which was displayed by immunofluorescence and western blot. In turn, delocalization of FIRRE from nucleus to cytoplasm is observed after the loss of PTBP1. The RNA-protein complex in the cytoplasm directly bound to BECN1 mRNA, and the binding site was at the 3' end of the mRNA. Cells with FIRRE and PTBP1 depletion alone or in combination were treated by Actinomycin D (ACD). Results of qPCR showed FIRRE stabilized BECN1 mRNA in a PTBP1-medieated manner. In addition, FIRRE contributed to autophagy activity. These findings indicate FIRRE acts as an oncogenic factor in CRC, which induces tumor development through stabilizing BECN1 mRNA and facilitating autophagy in a PTBP1-mediated manner.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Zhengyang Li
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Shizan Xu
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Wenjun Li
- Department of Anaesthesiology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201503, China
| | - Mengyun Chen
- General Practice of Huamu Community Health Service Center, 90 Yulan Road, Shanghai, 201204, China
| | - Miao Jiang
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China.
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China.
| |
Collapse
|
8
|
Wang Y, Li Z, Xu S, Li W, Chen M, Jiang M, Fan X. LncRNA FIRRE functions as a tumor promoter by interaction with PTBP1 to stabilize BECN1 mRNA and facilitate autophagy. Cell Death Dis 2022; 13:98. [PMID: 35110535 PMCID: PMC8811066 DOI: 10.1038/s41419-022-04509-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 12/02/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022]
Abstract
Long non-coding RNAs (lncRNAs) play critical functions in various cancers. Firre intergenic repeating RNA element (FIRRE), a lncRNA located in the nucleus, was overexpressed in colorectal cancer (CRC). However, the detailed mechanism of FIRRE in CRC remains elusive. Results of RNA sequence and qPCR illustrated overexpression of FIRRE in CRC cell lines and tissues. The aberrant expression of FIRRE was correlated with the migration, invasion, and proliferation in cell lines. In accordance, it was also associated with lymphatic metastasis and distant metastasis in patients with CRC. FIRRE was identified to physically interact with Polypyrimidine tract-binding protein (PTBP1) by RNA pull-down and RNA immunoprecipitation (RIP). Overexpression of FIRRE induced the translocation of PTBP1 from nucleus to cytoplasm, which was displayed by immunofluorescence and western blot. In turn, delocalization of FIRRE from nucleus to cytoplasm is observed after the loss of PTBP1. The RNA-protein complex in the cytoplasm directly bound to BECN1 mRNA, and the binding site was at the 3' end of the mRNA. Cells with FIRRE and PTBP1 depletion alone or in combination were treated by Actinomycin D (ACD). Results of qPCR showed FIRRE stabilized BECN1 mRNA in a PTBP1-medieated manner. In addition, FIRRE contributed to autophagy activity. These findings indicate FIRRE acts as an oncogenic factor in CRC, which induces tumor development through stabilizing BECN1 mRNA and facilitating autophagy in a PTBP1-mediated manner.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Zhengyang Li
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Shizan Xu
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China
| | - Wenjun Li
- Department of Anaesthesiology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201503, China
| | - Mengyun Chen
- General Practice of Huamu Community Health Service Center, 90 Yulan Road, Shanghai, 201204, China
| | - Miao Jiang
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China.
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201508, China.
| |
Collapse
|
9
|
Non-coding RNA-mediated autophagy in cancer: A protumor or antitumor factor? Biochim Biophys Acta Rev Cancer 2021; 1876:188642. [PMID: 34715268 DOI: 10.1016/j.bbcan.2021.188642] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022]
Abstract
Autophagy, usually referred to as macroautophagy, is a cytoprotective behavior that helps cells, especially cancer cells, escape crises. However, the role of autophagy in cancer remains controversial. The induction of autophagy is favorable for tumor growth, as it can degrade damaged cell components accumulated during nutrient deficiency, chemotherapy, or other stresses in a timely manner. Whereas the antitumor effect of autophagy might be closely related to its crosstalk with metabolism, immunomodulation, and other pathways. Recent studies have verified that lncRNAs and circRNAs modulate autophagy in carcinogenesis, cancer cells proliferation, apoptosis, metastasis, and chemoresistance via multiple mechanisms. A comprehensive understanding of the regulatory relationships between ncRNAs and autophagy in cancer might resolve chemoresistance and also offer intervention strategies for cancer therapy. This review systematically displays the regulatory effects of lncRNAs and circRNAs on autophagy in the contexts of cancer initiation, progression, and resistance to chemo- or radiotherapy and provides a novel insight into cancer therapy.
Collapse
|
10
|
Autophagy-Related Proteins Are Differentially Expressed in Adrenal Cortical Tumor/Pheochromocytoma and Associated with Patient Prognosis. Int J Mol Sci 2021; 22:ijms221910490. [PMID: 34638836 PMCID: PMC8508962 DOI: 10.3390/ijms221910490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
The aim of this research was to evaluate the expression and concomitant implications of LC3A, LC3B, beclin-1, and p62, which are key components of autophagy in human adrenal gland tumors. Tissue microarray was made for 321 cases of adrenal gland tumor (adrenal cortical adenoma (ACA): 115, adrenal cortical carcinoma (ACC): 17, and pheochromocytoma (PCC): 189). Immunohistochemical staining was performed for beclin-1, p62, LC3A, and LC3B, and the results were compared with the patients’ clinicopathologic parameters. LC3A, LC3B, beclin-1, and LC3B isolated single positive cells (ISPC) positivity rates were higher in PCC than in adrenal cortical tumor (ACT), whereas p62 positivity was lower in PCC than in ACT. The proportion of positive LC3B (ISPC) was higher in ACC than in ACA. In addition, the proportion of cells positive for p62 and LC3B (ISPC) was significantly higher in PCCs with a GAPP score of ≥3. In univariate Cox analysis, p62 positivity (p = 0.014) and the presence of p62 (ISPC) (p = 0.001) were associated with shorter disease-free survival in PCC. Moreover, p62 positivity was predictive of shorter overall survival (OS) in patients with PCC by multivariate analysis (relative risk, 6.240; 95% CI, 1.434–27.15; p = 0.015). Differences were found in the expression of autophagy-related proteins according to adrenal gland tumor types. Compared to ACT, the proportion of LC3A, LC3B, beclin-1, and LC3B (ISPC) positivity was higher in PCC, whereas p62 positivity was lower. Similarly, p62 positivity in PCC was associated with patient prognosis of OS.
Collapse
|
11
|
Autophagy Modulators in Cancer Therapy. Int J Mol Sci 2021; 22:ijms22115804. [PMID: 34071600 PMCID: PMC8199315 DOI: 10.3390/ijms22115804] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a process of self-degradation that plays an important role in removing damaged proteins, organelles or cellular fragments from the cell. Under stressful conditions such as hypoxia, nutrient deficiency or chemotherapy, this process can also become the strategy for cell survival. Autophagy can be nonselective or selective in removing specific organelles, ribosomes, and protein aggregates, although the complete mechanisms that regulate aspects of selective autophagy are not fully understood. This review summarizes the most recent research into understanding the different types and mechanisms of autophagy. The relationship between apoptosis and autophagy on the level of molecular regulation of the expression of selected proteins such as p53, Bcl-2/Beclin 1, p62, Atg proteins, and caspases was discussed. Intensive studies have revealed a whole range of novel compounds with an anticancer activity that inhibit or activate regulatory pathways involved in autophagy. We focused on the presentation of compounds strongly affecting the autophagy process, with particular emphasis on those that are undergoing clinical and preclinical cancer research. Moreover, the target points, adverse effects and therapeutic schemes of autophagy inhibitors and activators are presented.
Collapse
|
12
|
Zhao H, Shi L, Kong R, Li Z, Liu F, Zhao H, Li Z. Autophagy induction in tumor surrounding cells promotes tumor growth in adult Drosophila intestines. Dev Biol 2021; 476:294-307. [PMID: 33940033 DOI: 10.1016/j.ydbio.2021.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
During tumorigenesis, tumor cells interact intimately with their surrounding cells (microenvironment) for their growth and progression. However, the roles of tumor microenvironment in tumor development and progression are not fully understood. Here, using an established benign tumor model in adult Drosophila intestines, we find that non-cell autonomous autophagy (NAA) is induced in tumor surrounding neighbor cells. Tumor growth can be significantly suppressed by genetic ablation of autophagy induction in tumor neighboring cells, indicating that tumor neighboring cells act as tumor microenvironment to promote tumor growth. Autophagy in tumor neighboring cells is induced downstream of elevated ROS and activated JNK signaling in tumor cells. Interestingly, we find that active transport of nutrients, such as amino acids, from tumor neighboring cells sustains tumor growth, and increasing nutrient availability could significantly restore tumor growth. Together, these data demonstrate that tumor cells take advantage of their surrounding normal neighbor cells as nutrient sources through NAA to meet their high metabolic demand for growth and progression. Thus we provide insights into our understanding of the mechanisms underlying the interaction between tumor cells and their microenvironment in tumor development.
Collapse
Affiliation(s)
- Hang Zhao
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Lin Shi
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Ruiyan Kong
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Zhengran Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Fuli Liu
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Huiqing Zhao
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China.
| |
Collapse
|
13
|
Lanzillotta C, Di Domenico F. Stress Responses in Down Syndrome Neurodegeneration: State of the Art and Therapeutic Molecules. Biomolecules 2021; 11:biom11020266. [PMID: 33670211 PMCID: PMC7916967 DOI: 10.3390/biom11020266] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) is the most common genomic disorder characterized by the increased incidence of developing early Alzheimer’s disease (AD). In DS, the triplication of genes on chromosome 21 is intimately associated with the increase of AD pathological hallmarks and with the development of brain redox imbalance and aberrant proteostasis. Increasing evidence has recently shown that oxidative stress (OS), associated with mitochondrial dysfunction and with the failure of antioxidant responses (e.g., SOD1 and Nrf2), is an early signature of DS, promoting protein oxidation and the formation of toxic protein aggregates. In turn, systems involved in the surveillance of protein synthesis/folding/degradation mechanisms, such as the integrated stress response (ISR), the unfolded stress response (UPR), and autophagy, are impaired in DS, thus exacerbating brain damage. A number of pre-clinical and clinical studies have been applied to the context of DS with the aim of rescuing redox balance and proteostasis by boosting the antioxidant response and/or inducing the mechanisms of protein re-folding and clearance, and at final of reducing cognitive decline. So far, such therapeutic approaches demonstrated their efficacy in reverting several aspects of DS phenotype in murine models, however, additional studies aimed to translate these approaches in clinical practice are still needed.
Collapse
|
14
|
Lefort S, Balani S, Pellacani D, Guyot B, Gorski SM, Maguer-Satta V, Eaves CJ. Single-cell analysis of autophagy activity in normal and de novo transformed human mammary cells. Sci Rep 2020; 10:20266. [PMID: 33219251 PMCID: PMC7679376 DOI: 10.1038/s41598-020-77347-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022] Open
Abstract
Assessment of autophagy activity has historically been limited to investigations of fixed tissue or bulk cell populations. To address questions of heterogeneity and relate measurements to functional properties of viable cells isolated from primary tissue, we created a lentiviral (RFP-GFP-MAP1LC3B) vector that allows the autophagosome and autolysosome content of transduced cells to be monitored at the single-cell level. Use of this strategy to analyze purified subsets of normal human mammary cells showed that both the luminal progenitor-containing (LP) subset and the basal cells (BCs) display highly variable but overall similar autophagic flux activity despite differences suggested by measurements of the proteins responsible (i.e., LC3B, ATG7 and BECLIN1) in bulk lysates. Autophagosome content was also highly variable in the clonogenic cells within both the LPs and BCs, but the proliferative response of the BCs was more sensitive to autophagy inhibition. In addition, use of this vector showed cells with the lowest autophagosome content elicited the fastest tumor growth in 2 different models of human mammary tumorigenesis. These results illustrate the utility of this vector to define differences in the autophagy properties of individual cells in primary tissue and couple these with their responses to proliferative and oncogenic stimuli.
Collapse
Affiliation(s)
- Sylvain Lefort
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada. .,Centre de Recherche en Cancérologie de LyonInserm U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France.
| | - Sneha Balani
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Davide Pellacani
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Boris Guyot
- Centre de Recherche en Cancérologie de LyonInserm U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Sharon M Gorski
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada
| | - Véronique Maguer-Satta
- Centre de Recherche en Cancérologie de LyonInserm U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Connie J Eaves
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| |
Collapse
|
15
|
Agarwal S, Maekawa T. Nano delivery of natural substances as prospective autophagy modulators in glioblastoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102270. [PMID: 32702467 DOI: 10.1016/j.nano.2020.102270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the most destructive type of malignant brain tumor in humans due to cancer relapse. Latest studies have indicated that cancer cells are more reliant on autophagy for survival than non-cancer cells. Autophagy is entitled as programmed cell death type II and studies imply that it is a comeback of cancer cells to innumerable anti-cancer therapies. To diminish the adverse consequences of chemotherapeutics, numerous herbs of natural origin have been retained in cancer treatments. Additionally, autophagy induction occurs via their tumor suppressive actions that could cause cell senescence and increase apoptosis-independent cell death. However, most of the drugs have poor solubility and thus nano drug delivery systems possess excessive potential to improve the aqueous solubility and bioavailability of encapsulated drugs. There is a pronounced need for more therapies for glioblastoma treatment and hereby, the fundamental mechanisms of natural autophagy modulators in glioblastoma are prudently reviewed in this article.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan.
| | - Toru Maekawa
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
16
|
Martinez EF, de Araújo VC, Navarini NF, de Souza IF, Rena GB, Demasi APD, de Paula E, Teixeira LN. Microvesicles derived from squamous cell carcinoma induce cell death, autophagy, and invasion of benign myoepithelial cells. J Oral Pathol Med 2020; 49:761-770. [PMID: 32453894 DOI: 10.1111/jop.13037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND There has been great interest recently in the mechanisms of cell-to-cell communication through microvesicles (MV). These structures are produced by many different cell types and can modulate cellular activity by induction of epigenetic alterations. These vesicles may promote tumor mass increase either by stimulating cell proliferation via growth factors or by inhibiting apoptosis, which reinforces the role of such vesicles as important modulators of tumor progression. METHODS The present in vitro study aimed to characterize MV derived from malignant neoplastic epithelial cell cultures (EP) and their effect on the expression of apoptosis/autophagy and invasion related genes of benign myoepithelial (Myo) cell cultures. RESULTS The results revealed round structures with a mean size of 153.6 (±0.2) nm, with typical MV morphology. CD63 quantification indicated that EP cell culture at 70%-80% confluence secreted 3.088 × 108 MV/mL. Overall, Myo exposed to MVs derived from EP showed both up- and downregulation of tumorigenesis promoting genes. MVs from EP cells promoted cell death of Myo cells and positively modulate BAX, SURVIVIN, LC3B, MMP-2, and MMP-9 expression. Furthermore, an increasing of MMP-2 and MMP-9 secretion by Myo was observed after MV exposure. CONCLUSIONS These findings suggest that MVs from EP modulate autophagy of Myo cells, which may, in part, explain the disappearance of these cells in in situ areas of invasive carcinoma ex-pleomorphic adenoma. Additionally, the overexpression of MMPs contributes to the development of an invasive phenotype of Myo cells, which could favor the dissolution of the basement membrane during tumorigenesis process.
Collapse
Affiliation(s)
| | | | | | | | - Gabriel Bernardo Rena
- Cell Biology and Oral Pathology Division, Faculdade São Leopoldo Mandic, Campinas, SP, Brazil
| | - Ana Paula Dias Demasi
- Cell Biology and Oral Pathology Division, Faculdade São Leopoldo Mandic, Campinas, SP, Brazil
| | - Eneida de Paula
- Department of Biochemistry and Tissue Biology, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| | - Lucas Novaes Teixeira
- Cell Biology and Oral Pathology Division, Faculdade São Leopoldo Mandic, Campinas, SP, Brazil
| |
Collapse
|
17
|
Laskar YB, Mazumder PB. Insight into the molecular evidence supporting the remarkable chemotherapeutic potential of Hibiscus sabdariffa L. Biomed Pharmacother 2020; 127:110153. [PMID: 32344257 DOI: 10.1016/j.biopha.2020.110153] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 02/08/2023] Open
Abstract
Hibiscus sabdariffa or roselle tea is popular around the globe for its antioxidant capability along with various other health benefits. Besides, it has uses in Ayurvedic and Chinese herbal medicines for the treatment of several diseases. However, the investigation for the anticancer potential of the plant began roughly in the last decade that emerged with encouraging results. Both crude extracts and pure compounds of the plant were reported to induce chemoprevention, selective cytotoxicity, cell cycle arrest, apoptosis, autophagy and anti-metastasis effects in varied types of human cancer cells. The plant contains a high quantity of polyphenolic compounds and at least two of them were proven to induce potent anticancer effects. Although, the molecular mechanism underlying the anticancer activity was roughly estimated in several studies. The present review aimed to assemble all ambiguous information to report the molecular evidence establishing the potent anticancer activity of Hibiscus sabdariffa and its implication for cancer therapy. This study suggests that Hibiscus sabdariffa is an ideal candidate to investigate its role as a herbal supplement for cancer prevention and treatment. With excellent safety and tolerability record, polyphenolic compounds from the plant need better designed clinical trials.
Collapse
Affiliation(s)
- Yahyea Baktiar Laskar
- Natural Product & Biomedicine Research Laboratory, Department of Biotechnology, Assam University, Silchar, 788011, India
| | - Pranab Behari Mazumder
- Natural Product & Biomedicine Research Laboratory, Department of Biotechnology, Assam University, Silchar, 788011, India.
| |
Collapse
|
18
|
Chen Y, Li Q, Li Q, Xing S, Liu Y, Liu Y, Chen Y, Liu W, Feng F, Sun H. p62/SQSTM1, a Central but Unexploited Target: Advances in Its Physiological/Pathogenic Functions and Small Molecular Modulators. J Med Chem 2020; 63:10135-10157. [DOI: 10.1021/acs.jmedchem.9b02038] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Qi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Qihang Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yang Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People’s Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People’s Republic of China
| |
Collapse
|
19
|
Zhang C, Jiang J, Wang L, Zheng L, Xu J, Qi X, Huang H, Lu J, Li K, Wang H. Identification of Autophagy-Associated Biomarkers and Corresponding Regulatory Factors in the Progression of Colorectal Cancer. Front Genet 2020; 11:245. [PMID: 32265986 PMCID: PMC7100633 DOI: 10.3389/fgene.2020.00245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/28/2020] [Indexed: 12/30/2022] Open
Abstract
Autophagy is a self-degradation process that maintains homeostasis against stress in cells. Autophagy dysfunction plays a central role in the development of tumors, such as colorectal cancer (CRC). In this study, autophagy-related differentially expressed genes, their downstream functions, and upstream regulatory factors including RNA-binding proteins (RBP) involved in programmed cell death in the CRC were investigated. Transcription factors (TFs) and miRNAs have been shown to mainly regulate autophagy genes. Interestingly, we found that some of the RBP in the CRC, such as DDX17, SETDB1, and POLR3A, play an important regulatory role in maintaining autophagy at a basal level during growth by acting as TFs that regulate autophagy. Promoter methylations showed negative regulations on differentially expressed autophagy gene (DEAG), while copy number variations revealed a positive role in them. A proportional hazards regression analysis indicated that using autophagy-related prognostic signature can divide patients into high-risk and low-risk groups. Autophagy associated FDA-approved drugs were studied by a prognostic network. This would contribute to the identifications of new potential molecular therapeutic targets for CRC.
Collapse
Affiliation(s)
- Chunrui Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.,Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jing Jiang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liqiang Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Liyu Zheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jiankai Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiaolin Qi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education and College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Huiying Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jianping Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kongning Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education and College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Hong Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education and College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| |
Collapse
|
20
|
Tao M, Liu T, You Q, Jiang Z. p62 as a therapeutic target for tumor. Eur J Med Chem 2020; 193:112231. [PMID: 32193054 DOI: 10.1016/j.ejmech.2020.112231] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
p62/SQSTM1 (hereafter as p62) is a stress-inducible cellular protein, which interacts with various signaling proteins to regulate a variety of cellular functions. Growing lines of evidence supported a critical role of p62 in tumorigenesis, and p62 may become a therapeutic target for tumor. In this review, we summarize biological functions of structural domains of p62, reported bioactive molecules targeting p62, and the relationship between p62 and tumorigenesis.
Collapse
Affiliation(s)
- Mengmin Tao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tian Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhengyu Jiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
21
|
Zheng X, Jin X, Liu X, Liu B, Li P, Ye F, Zhao T, Chen W, Li Q. Inhibition of endoplasmic reticulum stress-induced autophagy promotes the killing effect of X-rays on sarcoma in mice. Biochem Biophys Res Commun 2019; 522:612-617. [PMID: 31785812 DOI: 10.1016/j.bbrc.2019.11.160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 11/22/2019] [Indexed: 12/30/2022]
Abstract
Endoplasmic reticulum (ER) stress is a conserved cellular process for cells to clear unfolded or misfolded proteins and maintain cell homeostasis under stress conditions. Autophagy may act as a pro-survival strategy to cope with multiple stress conditions in tumor progression and distant metastasis. Although many studies have demonstrated that there is a close correlation between radiation-induced ER stress and autophagy, the molecular mechanisms currently remain unclear. In the present study, we performed an in vivo study concerning the effect of autophagy induced by ER stress on the radiosensitivity of mouse sarcoma using X-rays. Our results documented that X-rays could induce ER stress in sarcoma and then autophagy was activated by unfolded protein response (UPR) through the IRE1-JNK-pBcl2-Beclin1 signaling axis. The induction of autophagy caused a decline in cell apoptosis while inhibiting the autophagy resulted in increased apoptosis and inhibition of tumor progression. Combined treatment of X-ray exposure and chloroquine increased ER stress-related apoptosis and enhanced the radiosensitivity of mouse sarcoma that was not sensitive to X-ray irradiation alone. Thus, our study indicates that inhibition of ER stress-induced autophagy might be a novel strategy to improve the efficacy of radiotherapy against radioresistant sarcoma.
Collapse
Affiliation(s)
- Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Bingtao Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China.
| |
Collapse
|
22
|
Ding X, Yue W, Chen H. Effect of artesunate on apoptosis and autophagy in tamoxifen resistant breast cancer cells (TAM-R). Transl Cancer Res 2019; 8:1863-1872. [PMID: 35116937 PMCID: PMC8797964 DOI: 10.21037/tcr.2019.08.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/12/2019] [Indexed: 11/24/2022]
Abstract
Background The antitumor effect of artesunate (ART) is well-recognized. To investigate the effect of ART on tamoxifen-resistant breast cancer cells (TAM-R) proliferation, apoptosis, and autophagy with TAM-R cells of breast cancer as objects of study, and to investigate whether ART could re-sensitize TAM-R cells to TAM therapy. Methods Experiments were performed using TAM-R cell lines. Cell Death Detection ELISA kit was used to detect the level of apoptosis. Western blot and immunofluorescent staining analysis were conducted to detect autophagy and apoptosis related proteins in TAM-R cells. Results After treated with ART, the proliferation activity of TAM-R cells was inhibited in a concentration-dependent manner. Increased apoptosis activity was detected in TAM-R cells when treated with ART. Compared with 10−6 M TAM monotherapy group, treatment group with ART and TAM in combination caused significant reduction in the levels of Bcl-2, XIAP, and Survivin proteins, and elevation of caspase-7. However, increased p53 proteins was not detected after ART treatment. No significant change was observed in autophagy proteins LC-3 and Beclin-1 among control, ART, TAM, and ART combined with TAM groups. Conclusions The intervention of ART could not inhibit protective autophagy in TAM-R cells, however, possess potential in inducing apoptosis. In addition, as ART inhibit TAM-R cells growth in a dose-dependent manner, co-administration of 1 or 3 µM of ART with 10−6 M TAM might not be enough to re-sensitize TAM-R cells to TAM therapy.
Collapse
Affiliation(s)
- Xiaoqing Ding
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| | - Wei Yue
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA 22908, USA
| | - Haiyan Chen
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| |
Collapse
|
23
|
Guo L, Li Y, Tian Y, Gong S, Chen X, Peng T, Wang A, Jiang Z. eIF2α promotes vascular remodeling via autophagy in monocrotaline-induced pulmonary arterial hypertension rats. Drug Des Devel Ther 2019; 13:2799-2809. [PMID: 31496656 PMCID: PMC6698179 DOI: 10.2147/dddt.s213817] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Eukaryotic initiation factor 2α (eIF2α) plays important roles in the proliferation and survival of pulmonary artery smooth muscle cells (PASMCs) in animal hypoxia-induced pulmonary hypertension models. However, the underlying mechanism remains unknown at large. Autophagy has been reported to play a key role in the vascular remodeling in pulmonary arterial hypertension (PAH). The purposes of this study are to determine the functions of eIF2α and autophagy in the vascular remodeling of the monocrotaline-induced PAH rats and to clarify the correlation between eIF2α and autophagy. METHODS We established a rat model of monocrotaline-induced PAH, and we established a cell model of platelet derived growth factor (PDGF)-induced PASMCs proliferation. The vascular morphology and the expression of eIF2α, LC3B, and p62 were assessed in the pulmonary arterial tissue of Sprague-Dawleyrats and PDGF-induced PASMCs. RESULTS Autophagy was significantly active in monocrotaline model group (MCT)-induced PAH rats, which obviously promotes vascular remodeling in MCT-induced PAH rats. Furthermore, the proliferation of PASMCs was induced by PDGF in vitro. The expression of LC3B, eIF2α was increased in the PDGF-induced PASMCs proliferation, and the expression of p62 was reduced in the PDGF-induced PASMCs proliferation. Moreover, eIF2α siRNA downregulated the expression of eIF2α and LC3B, and upregulated the expression of p62 in PDGF-induced PASMCs proliferation. eIF2α siRNA inhibited the PDGF-induced PASMCs proliferation. Finally, chloroquine can upregulate the protein expression of LC3B and p62, it also can inhibit proliferation in PDGF-induced PASMCs. CONCLUSION Based on these observations, we conclude that eIF2α promotes the proliferation of PASMCs and vascular remodeling in monocrotaline-induced PAH rats through accelerating autophagy pathway.
Collapse
Affiliation(s)
- Linya Guo
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, Hengyang421001, People’s Republic of China
| | - Yanbing Li
- National Key Discipline of Human Anatomy, Southern Medical University, Guangzhou510000, Guangdong, People’s Republic of China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangzhou, 510000, Guangdong, People’s Republic of China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang421002, Hunan, People’s Republic of China
- Postdoctoral Research Institute on Basic Medicine, University of South China, Hengyang, 421001, Hunan, People’s Republic of China
| | - Shaoxin Gong
- Department of Pathology, First Affiliated Hospital, University of South China, Hengyang421001, Hunan, People’s Republic of China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, Hengyang421001, People’s Republic of China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, Hengyang421001, People’s Republic of China
| | - Aiping Wang
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, Hengyang421001, People’s Republic of China
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang421002, Hunan, People’s Republic of China
- Postdoctoral Research Institute on Basic Medicine, University of South China, Hengyang, 421001, Hunan, People’s Republic of China
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang421001, Hunan, People’s Republic of China
| | - Zhisheng Jiang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang421001, Hunan, People’s Republic of China
| |
Collapse
|
24
|
Cui L, Wang X, Zhao X, Kong C, Li Z, Liu Y, Jiang X, Zhang X. The autophagy-related genes Beclin1 and LC3 in the prognosis of pancreatic cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2989-2996. [PMID: 31934136 PMCID: PMC6949699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/25/2019] [Indexed: 06/10/2023]
Abstract
PURPOSE To investigate the role of the autophagy-related genes Beclin1 and LC3 in the prognosis of pancreatic cancer. METHODS A total of 86 pancreatic cancer tissues and 84 paired, adjacent normal pancreatic tissues were collected from 86 patients who underwent pancreatic resection surgery in our hospital from January 2009 to August 2011. Demographic data including age, gender, family cancer history, and clinic-pathological characteristics, including tumor diameter, differential, TNM staging and lymphatic metastasis were collected. The expressions of Beclin1 and LC3 were determined using both immunohistochemistry (IHC) and RT-qPCR. RESULTS The expression levels of both Beclin1 and LC3 mRNA and proteins were significantly up-regulated in the tumor tissues compared with the normal tissues. Higher expressions of Beclin1 and LC3 were found in the tumor tissues of patients with TNM stages III~IV, patients with lymphatic metastasis, and patients who died. Meanwhile Beclin1 and LC3 correlated with TNM stage, differential condition, and the patients' lymphatic metastasis rates. A survival analysis showed that patients with low expressions of Beclin1 and LC3 had longer survival times, and both the Beclin1 and LC3 genes were independent risk factors for 5-year mortality in pancreatic cancer patients. CONCLUSION The Beclin1 and LC3 genes correlate with the tumor stage, metastasis conditions, and pancreatic cancer patients' mortality.
Collapse
Affiliation(s)
- Long Cui
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Xiaochuan Wang
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Xin Zhao
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Chenchen Kong
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Zhengchen Li
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Yangsui Liu
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Xinchun Jiang
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| | - Xinhui Zhang
- Department of Hepatopancreatobiliary Surgery, Xuzhou Central Hospital Affiliated to Xuzhou Medical University Xuzhou, Jiangsu Province, China
| |
Collapse
|
25
|
Granular Cell Tumor in a Horse: Multifocal Pulmonary Distribution and Evidence of Autophagy in Tumorigenesis. J Equine Vet Sci 2019; 79:23-29. [PMID: 31405495 DOI: 10.1016/j.jevs.2019.05.010] [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: 02/12/2019] [Revised: 03/27/2019] [Accepted: 05/09/2019] [Indexed: 11/20/2022]
Abstract
Granular cell tumor (GCT) is a soft tissue neoplasm characterized by abundant intracellular eosinophilic granules. The majority of GCTs are benign, although some display malignant behavior. Furthermore, GCTs may mimic other neoplasms. The clinical course and biology of GCTs are poorly understood. Regarding the histogenesis of GCT, a Schwann cell origin is currently favored in light of immunohistochemical and ultrastructural analyses. However, based on literature data, some of the primitive GCTs show non-neural origin; therefore, the histogenesis of this tumor has remained enigmatic. Granular cell tumors can arise in almost any location of the body and typically present as solitary lesions. This study illustrates equine primary GCT with multifocal pulmonary distribution. The presence of GCT in the respiratory tract becomes a diagnostic challenge on initial presentation. The morphologic details of this case are presented. Immunohistochemical evaluation confirmed the neuronal origin of equine GCT and the relation of intracytoplasmic granules formation to an autophagy phenomenon. Most of the discussion is related to GCT nature to help characterize molecular aspects associated with the biological behavior of this tumor and its heterogeneity.
Collapse
|
26
|
Skarkova V, Kralova V, Vitovcova B, Rudolf E. Selected Aspects of Chemoresistance Mechanisms in Colorectal Carcinoma-A Focus on Epithelial-to-Mesenchymal Transition, Autophagy, and Apoptosis. Cells 2019; 8:cells8030234. [PMID: 30871055 PMCID: PMC6468859 DOI: 10.3390/cells8030234] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 12/15/2022] Open
Abstract
Chemoresistance has been found in all malignant tumors including colorectal carcinoma (CRC). Nowadays chemoresistance is understood as a major reason for therapy failure, with consequent tumor growth and spreading leading ultimately to the patient's premature death. The chemotherapy-related resistance of malignant colonocytes may be manifested in diverse mechanisms that may exist both prior to the onset of the therapy or after it. The ultimate function of this chemoresistance is to ensure the survival of malignant cells through continuing adaptation within an organism, therefore, the nature and spectrum of cell-survival strategies in CRC represent a highly significant target of scientific inquiry. Among these survival strategies employed by CRC cells, three unique but significantly linked phenomena stand out-epithelial-to-mesenchymal transition (EMT), autophagy, and cell death. In this mini-review, current knowledge concerning all three mechanisms including their emergence, timeline, regulation, and mutual relationships will be presented and discussed.
Collapse
Affiliation(s)
- Veronika Skarkova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Vera Kralova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Barbora Vitovcova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| |
Collapse
|
27
|
Jacomin AC, Gul L, Sudhakar P, Korcsmaros T, Nezis IP. What We Learned From Big Data for Autophagy Research. Front Cell Dev Biol 2018; 6:92. [PMID: 30175097 PMCID: PMC6107789 DOI: 10.3389/fcell.2018.00092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/27/2018] [Indexed: 12/13/2022] Open
Abstract
Autophagy is the process by which cytoplasmic components are engulfed in double-membraned vesicles before being delivered to the lysosome to be degraded. Defective autophagy has been linked to a vast array of human pathologies. The molecular mechanism of the autophagic machinery is well-described and has been extensively investigated. However, understanding the global organization of the autophagy system and its integration with other cellular processes remains a challenge. To this end, various bioinformatics and network biology approaches have been developed by researchers in the last few years. Recently, large-scale multi-omics approaches (like genomics, transcriptomics, proteomics, lipidomics, and metabolomics) have been developed and carried out specifically focusing on autophagy, and generating multi-scale data on the related components. In this review, we outline recent applications of in silico investigations and big data analyses of the autophagy process in various biological systems.
Collapse
Affiliation(s)
| | - Lejla Gul
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - Padhmanand Sudhakar
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute, Norwich Research Park, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
- Gut Microbes and Health Programme, Quadram Institute, Norwich Research Park, Norwich, United Kingdom
| | - Ioannis P. Nezis
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
28
|
Pierzynowska K, Gaffke L, Cyske Z, Puchalski M, Rintz E, Bartkowski M, Osiadły M, Pierzynowski M, Mantej J, Piotrowska E, Węgrzyn G. Autophagy stimulation as a promising approach in treatment of neurodegenerative diseases. Metab Brain Dis 2018; 33:989-1008. [PMID: 29542037 PMCID: PMC6060747 DOI: 10.1007/s11011-018-0214-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
Autophagy is a process of degradation of macromolecules in the cytoplasm, particularly proteins of a long half-life, as well as whole organelles, in eukaryotic cells. Lysosomes play crucial roles during this degradation. Autophagy is a phylogenetically old, and evolutionarily conserved phenomenon which occurs in all eukaryotic cells. It can be found in yeast Saccharomyces cerevisiae, insect Drosophila melanogaster, and mammals, including humans. Its high importance for cell physiology has been recognized, and in fact, dysfunctions causing impaired autophagy are associated with many severe disorders, including cancer and metabolic brain diseases. The types and molecular mechanisms of autophagy have been reviewed recently by others, and in this paper they will be summarized only briefly. Regulatory networks controlling the autophagy process are usually described as negative regulations. In contrast, here, we focus on different ways by which autophagy can be stimulated. In fact, activation of this process by different factors or processes can be considered as a therapeutic strategy in metabolic neurodegenerative diseases. These aspects are reviewed and discussed in this article.
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Zuzanna Cyske
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Michał Puchalski
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Michał Bartkowski
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Marta Osiadły
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Michał Pierzynowski
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Jagoda Mantej
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Ewa Piotrowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland.
| |
Collapse
|
29
|
Giorgi C, Marchi S, Simoes IC, Ren Z, Morciano G, Perrone M, Patalas-Krawczyk P, Borchard S, Jȩdrak P, Pierzynowska K, Szymański J, Wang DQ, Portincasa P, Wȩgrzyn G, Zischka H, Dobrzyn P, Bonora M, Duszynski J, Rimessi A, Karkucinska-Wieckowska A, Dobrzyn A, Szabadkai G, Zavan B, Oliveira PJ, Sardao VA, Pinton P, Wieckowski MR. Mitochondria and Reactive Oxygen Species in Aging and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:209-344. [PMID: 30072092 PMCID: PMC8127332 DOI: 10.1016/bs.ircmb.2018.05.006] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aging has been linked to several degenerative processes that, through the accumulation of molecular and cellular damage, can progressively lead to cell dysfunction and organ failure. Human aging is linked with a higher risk for individuals to develop cancer, neurodegenerative, cardiovascular, and metabolic disorders. The understanding of the molecular basis of aging and associated diseases has been one major challenge of scientific research over the last decades. Mitochondria, the center of oxidative metabolism and principal site of reactive oxygen species (ROS) production, are crucial both in health and in pathogenesis of many diseases. Redox signaling is important for the modulation of cell functions and several studies indicate a dual role for ROS in cell physiology. In fact, high concentrations of ROS are pathogenic and can cause severe damage to cell and organelle membranes, DNA, and proteins. On the other hand, moderate amounts of ROS are essential for the maintenance of several biological processes, including gene expression. In this review, we provide an update regarding the key roles of ROS-mitochondria cross talk in different fundamental physiological or pathological situations accompanying aging and highlighting that mitochondrial ROS may be a decisive target in clinical practice.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Ines C.M. Simoes
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Giampaolo Morciano
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Maria Pia Hospital, GVM Care & Research, Torino, Italy
| | - Mariasole Perrone
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paulina Patalas-Krawczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sabine Borchard
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Paulina Jȩdrak
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | - Jȩdrzej Szymański
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - David Q. Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Dept. of Biomedical Sciences & Human Oncology, University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Grzegorz Wȩgrzyn
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Pawel Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Massimo Bonora
- Departments of Cell Biology and Gottesman Institute for Stem Cell & Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jerzy Duszynski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Alessandro Rimessi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | | | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Barbara Zavan
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Paulo J. Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Vilma A. Sardao
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Paolo Pinton
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
| | - Mariusz R. Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
30
|
Alizadeh J, Glogowska A, Thliveris J, Kalantari F, Shojaei S, Hombach-Klonisch S, Klonisch T, Ghavami S. Autophagy modulates transforming growth factor beta 1 induced epithelial to mesenchymal transition in non-small cell lung cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:749-768. [PMID: 29481833 DOI: 10.1016/j.bbamcr.2018.02.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/15/2018] [Accepted: 02/18/2018] [Indexed: 02/07/2023]
Abstract
Lung cancer is considered one of the most frequent causes of cancer-related death worldwide and Non-Small Cell Lung Cancer (NSCLC) accounts for 80% of all lung cancer cases. Autophagy is a cellular process responsible for the recycling of damaged organelles and protein aggregates. Transforming growth factor beta-1 (TGFβ1) is involved in Epithelial to Mesenchymal Transition (EMT) and autophagy induction in different cancer models and plays an important role in the pathogenesis of NSCLC. It is not clear how autophagy can regulate EMT in NSCLC cells. In the present study, we have investigated the regulatory role of autophagy in EMT induction in NSCLC and show that TGFβ1 can simultaneously induce both autophagy and EMT in the NSCL lines A549 and H1975. Upon chemical inhibition of autophagy using Bafilomycin-A1, the expression of the mesenchymal marker vimentin and N-cadherin was reduced. Immunoblotting and immunocytochemistry (ICC) showed that the mesenchymal marker vimentin was significantly downregulated upon TGFβ1 treatment in ATG7 knockdown cells when compared to corresponding cells treated with scramble shRNA (negative control), while E-cadherin was unchanged. Furthermore, autophagy inhibition (Bafilomycin A1 and ATG7 knockdown) decreased two important mesenchymal functions, migration and contraction, of NSCLC cells upon TGFβ1 treatment. This study identified a crucial role of autophagy as a potential positive regulator of TGFβ1-induced EMT in NSCLC cells and identifies inhibitors of autophagy as promising new drugs in antagonizing the role of EMT inducers, like TGFβ1, in the clinical progression of NSCLC.
Collapse
Affiliation(s)
- Javad Alizadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Aleksandra Glogowska
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - James Thliveris
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Forouh Kalantari
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Medical Microbiology & Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada; Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
31
|
Dual role of autophagy in hallmarks of cancer. Oncogene 2017; 37:1142-1158. [PMID: 29255248 DOI: 10.1038/s41388-017-0046-6] [Citation(s) in RCA: 392] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/09/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
Evolutionarily conserved across eukaryotic cells, macroautophagy (herein autophagy) is an intracellular catabolic degradative process targeting damaged and superfluous cellular proteins, organelles, and other cytoplasmic components. Mechanistically, it involves formation of double-membrane vesicles called autophagosomes that capture cytosolic cargo and deliver it to lysosomes, wherein the breakdown products are eventually recycled back to the cytoplasm. Dysregulation of autophagy often results in various disease manifestations, including neurodegeneration, microbial infections, and cancer. In the case of cancer, extensive attention has been devoted to understanding the paradoxical roles of autophagy in tumor suppression and tumor promotion. In this review, while we summarize how this self-eating process is implicated at various stages of tumorigenesis, most importantly, we address the link between autophagy and hallmarks of cancer. This would eventually provide a better understanding of tumor dependence on autophagy. We also discuss how therapeutics targeting autophagy can counter various transformations involved in tumorigenesis. Finally, this review will provide a novel insight into the mutational landscapes of autophagy-related genes in several human cancers, using genetic information collected from an array of cancers.
Collapse
|
32
|
Monitoring Autophagy Immunohistochemically and Ultrastructurally during Human Head and Neck Carcinogenesis. Relationship with the DNA Damage Response Pathway. Int J Mol Sci 2017; 18:ijms18091920. [PMID: 28880214 PMCID: PMC5618569 DOI: 10.3390/ijms18091920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a catabolic process that preserves cellular homeostasis. Its exact role during carcinogenesis is not completely defined. Specifically in head and neck cancer, such information from clinical settings that comprise the whole spectrum of human carcinogenesis is very limited. Towards this direction, we examined the in situ status of the autophagy-related factors, Beclin-1, microtubule-associated protein 1 light chain 3, member B (LC3B) and sequestosome 1/p62 (p62) in clinical material covering all histopathological stages of human head and neck carcinogenesis. This material is unique as each panel of lesions is derived from the same patient and moreover we have previously assessed it for the DNA damage response (DDR) activation status. Since Beclin-1, LC3B and p62 reflect the nucleation, elongation and degradation stages of autophagy, respectively, their combined immunohistochemical (IHC) expression profiles could grossly mirror the autophagic flux. This experimental approach was further corroborated by ultrastructural analysis, applying transmission electron microscopy (TEM). The observed Beclin-1/LC3B/p62 IHC patterns, obtained from serial sections analysis, along with TEM findings are suggestive of a declined authophagic activity in preneoplastic lesions that was restored in full blown cancers. Correlating these findings with DDR status in the same pathological stages are indicative of: (i) an antitumor function of autophagy in support to that of DDR, possibly through energy deprivation in preneoplastic stages, thus preventing incipient cancer cells from evolving; and (ii) a tumor-supporting role in the cancerous stage.
Collapse
|
33
|
Expression of Autophagy-Related Proteins in Hürthle Cell Neoplasm Is Different from That in Follicular Neoplasm. DISEASE MARKERS 2017; 2017:1372387. [PMID: 28819333 PMCID: PMC5551561 DOI: 10.1155/2017/1372387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022]
Abstract
Purpose We aimed to evaluate expression of autophagy-related proteins in Hürthle cell neoplasm (HCN) and follicular neoplasm (FN) and assess the clinical implications. Methods 265 FNs (112 follicular carcinomas and 153 follicular adenomas) and 108 HCNs (27 Hürthle cell carcinomas and 81 Hürthle cell adenomas) were made into a tissue microarray. Immunohistochemical staining and Western blot for autophagy-related proteins (beclin-1, light chain (LC) 3A, LC3B, p62, and BNIP3) were performed, and the results were statistically analyzed. Results A higher expression rate of beclin-1, LC3B, p62, and BNIP3 was found in HCN than in FN (P < 0.001). The expression rate of beclin-1, LC3B, p62, and BNIP3 was the highest in HCCs followed by HCAs, FCs, and FAs in that order (P < 0.001). HCCs were positive for the largest number of autophagy-related proteins followed by HCAs, FCs, and FAs (P < 0.001), and most of the positive markers identified in HCCs were the high autophagy type (P < 0.001), defined by positive staining for three or more of the five autophagy-related proteins. Conclusion The autophagy-related proteins, beclin-1, LC3A, LC3B, p62, and BNIP3, were more frequently expressed in HCNs than in FNs, and HCCs showed the highest expression rate.
Collapse
|
34
|
Mathai BJ, Meijer AH, Simonsen A. Studying Autophagy in Zebrafish. Cells 2017; 6:E21. [PMID: 28698482 PMCID: PMC5617967 DOI: 10.3390/cells6030021] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/01/2017] [Accepted: 07/03/2017] [Indexed: 12/26/2022] Open
Abstract
Autophagy is an evolutionarily conserved catabolic process which allows lysosomal degradation of complex cytoplasmic components into basic biomolecules that are recycled for further cellular use. Autophagy is critical for cellular homeostasis and for degradation of misfolded proteins and damaged organelles as well as intracellular pathogens. The role of autophagy in protection against age-related diseases and a plethora of other diseases is now coming to light; assisted by several divergent eukaryotic model systems ranging from yeast to mice. We here give an overview of different methods used to analyse autophagy in zebrafish-a relatively new model for studying autophagy-and briefly discuss what has been done so far and possible future directions.
Collapse
Affiliation(s)
- Benan John Mathai
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, 0317 Oslo, Norway.
| | - Annemarie H Meijer
- Institute of Biology Leiden, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, 0317 Oslo, Norway.
| |
Collapse
|
35
|
Tong T, Niu YH, Yue Y, Wu SC, Ding H. Beneficial effects of anthocyanins from red cabbage (Brassica oleracea L. var. capitata L.) administration to prevent irinotecan-induced mucositis. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.01.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
36
|
Qi Y, Ruan J, Wang M, Dai Y, Zhou Q, Gui S, Zhang S, Wang Y. Effects of hyperbaric oxygen treatment on gastric cancer cell line SGC7901. Biomed Rep 2017; 6:475-479. [PMID: 28413648 DOI: 10.3892/br.2017.869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/14/2017] [Indexed: 01/02/2023] Open
Abstract
Hyperbaric oxygen (HBO) has been previously identified as an effective adjunct treatment option for the management of brain injury, diabetic ulcers and chronic wounds. However, the roles of HBO as an adjunctive therapy for tumors remain controversial. The present research project was performed to explore the effects of HBO treatment on proliferation, autophagy and endoplasmic reticulum stress response of the gastric cancer cell line, SGC7901. The present study demonstrated that, after subjecting SGC7901 cells to HBO treatment, the increase in cell proliferation was significant, compared with that of the control group. In addition, there was a significant increase in LC3-phosphatidylethanolamine conjugate (LC3-II) level, as well as binding immunoglobulin protein level, and a significant decrease in CCAAT-enhancer-binding protein homologous protein level. These suggested that hyperbaric oxygen treatment alone may promote proliferation and cell survival of gastric cancer cell SGC7901, and inhibit apoptosis through regulating cell autophagy and oxidative stress.
Collapse
Affiliation(s)
- Yinliang Qi
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Jianfeng Ruan
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Mei Wang
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Yuanchang Dai
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Qing Zhou
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shuyu Gui
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230011, P.R. China
| | - Sumei Zhang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
37
|
Kim HM, Kim ES, Koo JS. Expression of Autophagy-Related Proteins in Different Types of Thyroid Cancer. Int J Mol Sci 2017; 18:ijms18030540. [PMID: 28257096 PMCID: PMC5372556 DOI: 10.3390/ijms18030540] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 02/08/2023] Open
Abstract
Thyroid cancer is common type of malignant tumor in humans, and the autophagy status of such tumors may vary according to subtype. This study aimed to investigate the expression and implications of the major autophagy-related molecules light chain (LC) 3A, LC3B, p62, and BNIP-3 in human thyroid carcinoma. Tissue microarrays were constructed from 555 thyroid cancers (papillary thyroid carcinoma (PTC): 342; follicular carcinoma (FC): 112; medullary carcinoma (MC): 70; poorly differentiated carcinoma (PDC): 23; and anaplastic carcinoma (AC): 8) and 152 follicular adenomas (FAs). Expression of autophagy-related molecules (LC3A, LC3B, p62, and BNIP-3) was detected immunohistochemically, and the results were analyzed via comparison with clinicopathologic parameters. Tumoral LC3A and LC3B expressions were the highest in MC (p < 0.001), whereas stromal LC3A expression was higher in AC and PTC (p < 0.001). BNIP-3 expression was absent in MC and AC (p = 0.013). Tumoral LC3A, LC3B, and p62 expressions were higher in conventional type PTC, compared with those in the follicular variant. PTC with the BRAF V600E mutation exhibited higher expression of all autophagy-related proteins relative to PTC without this mutation (p < 0.05). BNIP-3 negativity was associated with capsular invasion in FC (p = 0.001), and p62 negativity was associated with lymph node metastasis in MC (p = 0.006). In a univariate analysis, LC3B negativity was associated with shorter disease-free survival in PTC with the BRAF V600E mutation (p = 0.024). We conclude that the expression of autophagy-related proteins differs according to thyroid cancer subtype.
Collapse
Affiliation(s)
- Hye Min Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 130-701, Korea.
| | - Eun-Sol Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 130-701, Korea.
| | - Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Seoul 130-701, Korea.
| |
Collapse
|
38
|
Mokarram P, Albokashy M, Zarghooni M, Moosavi MA, Sepehri Z, Chen QM, Hudecki A, Sargazi A, Alizadeh J, Moghadam AR, Hashemi M, Movassagh H, Klonisch T, Owji AA, Łos MJ, Ghavami S. New frontiers in the treatment of colorectal cancer: Autophagy and the unfolded protein response as promising targets. Autophagy 2017; 13:781-819. [PMID: 28358273 PMCID: PMC5446063 DOI: 10.1080/15548627.2017.1290751] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC), despite numerous therapeutic and screening attempts, still remains a major life-threatening malignancy. CRC etiology entails both genetic and environmental factors. Macroautophagy/autophagy and the unfolded protein response (UPR) are fundamental mechanisms involved in the regulation of cellular responses to environmental and genetic stresses. Both pathways are interconnected and regulate cellular responses to apoptotic stimuli. In this review, we address the epidemiology and risk factors of CRC, including genetic mutations leading to the occurrence of the disease. Next, we discuss mutations of genes related to autophagy and the UPR in CRC. Then, we discuss how autophagy and the UPR are involved in the regulation of CRC and how they associate with obesity and inflammatory responses in CRC. Finally, we provide perspectives for the modulation of autophagy and the UPR as new therapeutic options for CRC treatment.
Collapse
Affiliation(s)
- Pooneh Mokarram
- a Colorectal Research Center and Department of Biochemistry , School of Medicine, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mohammed Albokashy
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Maryam Zarghooni
- c Zabol University of Medical Sciences , Zabol , Iran.,d University of Toronto Alumni , Toronto , ON , Canada
| | - Mohammad Amin Moosavi
- e Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology , Tehran , Iran
| | - Zahra Sepehri
- c Zabol University of Medical Sciences , Zabol , Iran
| | - Qi Min Chen
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | | | | | - Javad Alizadeh
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Adel Rezaei Moghadam
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Mohammad Hashemi
- g Department of Clinical Biochemistry , School of Medicine, Zahedan University of Medical Sciences , Zahedan , Iran
| | - Hesam Movassagh
- h Department of Immunology , Rady Faculty of Health Sciences, College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Thomas Klonisch
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Ali Akbar Owji
- i Department of Clinical Biochemistry , School of Medicine, Shiraz Medical University , Shiraz , Iran
| | - Marek J Łos
- j Małopolska Centre of Biotechnology , Jagiellonian University , Krakow , Poland ; LinkoCare Life Sciences AB , Sweden
| | - Saeid Ghavami
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada.,k Health Policy Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
39
|
Zheng X, Jin X, Li F, Liu X, Liu Y, Ye F, Li P, Zhao T, Li Q. Inhibiting autophagy with chloroquine enhances the anti-tumor effect of high-LET carbon ions via ER stress-related apoptosis. Med Oncol 2017; 34:25. [PMID: 28070729 DOI: 10.1007/s12032-017-0883-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022]
Abstract
Energetic carbon ions (CI) offer great advantages over conventional radiations such as X- or γ-rays in cancer radiotherapy. High linear energy transfer (LET) CI can induce both endoplasmic reticulum (ER) stress and autophagy in tumor cells under certain circumstances. The molecular connection between ER stress and autophagy in tumor exposed to high-LET radiation and how these two pathways influence the therapeutic effect against tumor remain poorly understood. In this work, we studied the impact of autophagy and apoptosis induced by ER stress following high-LET CI radiation on the radiosensitivity of S180 cells both in vitro and in vivo. In the in vitro experiment, X-rays were also used as a reference radiation. Our results documented that the combination of CI radiation with chloroquine (CQ), a special autophagy inhibitor, produced more pronounced proliferation suppression in S180 cells and xenograft tumors. Co-treatment with CI radiation and CQ could block autophagy through the IRE1/JNK/Beclin-1 axis and enhance apoptotic cell death via the activation of C/EBP homologous protein (CHOP) by the IRE1 pathway rather than PERK in vitro and in vivo. Thus, our study indicates that inhibiting autophagy might be a promising therapeutic strategy in CI radiotherapy via aggravating the ER stress-related apoptosis.
Collapse
Affiliation(s)
- Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China. .,Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China. .,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
40
|
Functional and Molecular Insights of Hydrogen Sulfide Signaling and Protein Sulfhydration. J Mol Biol 2016; 429:543-561. [PMID: 28013031 DOI: 10.1016/j.jmb.2016.12.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S), a novel gasotransmitter, is endogenously synthesized by multiple enzymes that are differentially expressed in the peripheral tissues and central nervous systems. H2S regulates a wide range of physiological processes, namely cardiovascular, neuronal, immune, respiratory, gastrointestinal, liver, and endocrine systems, by influencing cellular signaling pathways and sulfhydration of target proteins. This review focuses on the recent progress made in H2S signaling that affects mechanistic and functional aspects of several biological processes such as autophagy, inflammation, proliferation and differentiation of stem cell, cell survival/death, and cellular metabolism under both physiological and pathological conditions. Moreover, we highlighted the cross-talk between nitric oxide and H2S in several bilogical contexts.
Collapse
|
41
|
Kunanopparat A, Kimkong I, Palaga T, Tangkijvanich P, Sirichindakul B, Hirankarn N. Increased ATG5-ATG12 in hepatitis B virus-associated hepatocellular carcinoma and their role in apoptosis. World J Gastroenterol 2016; 22:8361-8374. [PMID: 27729742 PMCID: PMC5055866 DOI: 10.3748/wjg.v22.i37.8361] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/04/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate autophagy-related genes, particularly ATG12, in apoptosis and cell cycle in hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) and non-HBV-HCC cell lines.
METHODS The expression of autophagy-related genes in HBV-associated hepatocellular carcinoma and non-HBV-HCC cell lines and human liver tissues was examined by quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR) and western blotting. The silencing of target genes was used to examine the function of various genes in apoptosis and cell cycle progression.
RESULTS The expression of autophagy related genes ATG5, ATG12, ATG9A and ATG4B expression was analyzed in HepG2.2.15 cells and compared with HepG2 and THLE cells. We found that ATG5 and ATG12 mRNA expression was significantly increased in HepG2.2.15 cells compared to HepG2 cells (P < 0.005). Moreover, ATG5-ATG12 protein levels were increased in tumor liver tissues compared to adjacent non-tumor tissues mainly from HCC patients with HBV infection. We also analyzed the function of ATG12 in cell apoptosis and cell cycle progression. The percentage of apoptotic cells increased by 11.4% in ATG12-silenced HepG2.2.15 cells (P < 0.005) but did not change in ATG12-silenced HepG2 cells under starvation with Earle’s balanced salt solution. However, the combination blockade of Notch signaling and ATG12 decreased the apoptotic rate of HepG2.2.15 cells from 55.6% to 50.4% (P < 0.05).
CONCLUSION ATG12 is important for HBV-associated apoptosis and a potential drug target for HBV-HCC. Combination inhibition of ATG12/Notch signaling had no additional effect on HepG2.2.15 apoptosis.
Collapse
|
42
|
Li YI, Libby EF, Lewis MJ, Liu J, Shacka JJ, Hurst DR. Increased autophagic response in a population of metastatic breast cancer cells. Oncol Lett 2016; 12:523-529. [PMID: 27347175 PMCID: PMC4906619 DOI: 10.3892/ol.2016.4613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 04/29/2016] [Indexed: 02/05/2023] Open
Abstract
Breast cancer cells are heterogeneous in their ability to invade and fully metastasize, and thus also in their capacity to survive the numerous stresses encountered throughout the multiple steps of the metastatic cascade. Considering the role of autophagy as a survival response to stress, the present study hypothesized that distinct populations of breast cancer cells may possess an altered autophagic capacity that influences their metastatic potential. It was observed that a metastatic breast cancer cell line, MDA-MB-231, that was sensitive to autophagic induction additionally possessed the ability to proliferate following nutrient deprivation. Furthermore, a selected subpopulation of these cells that survived multiple exposures to starvation conditions demonstrated a heightened response to autophagic induction compared to their parent cells. Although this subpopulation maintained a more grape-like pattern in three-dimensional culture compared to the extended spikes of the parent population, autophagic induction in this subpopulation elicited an invasive phenotype with extended spikes. Taken together, these results suggest that autophagic induction may contribute to the ability of distinct breast cancer cell populations to survive and invade.
Collapse
Affiliation(s)
- Y I Li
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Emily Falk Libby
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Monica J Lewis
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Jianzhong Liu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - John J Shacka
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Douglas R Hurst
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| |
Collapse
|
43
|
Koo JS, Kim JW, Yoon JS. Expression of Autophagy and Reactive Oxygen Species-Related Proteins in Lacrimal Gland Adenoid Cystic Carcinoma. Yonsei Med J 2016; 57:482-9. [PMID: 26847304 PMCID: PMC4740544 DOI: 10.3349/ymj.2016.57.2.482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/03/2015] [Accepted: 06/19/2015] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To investigate the difference of expression of autophagy and reactive oxygen species (ROS) related proteins in adenoid cystic carcinoma (ACC) of lacrimal gland in comparison with ACC of salivary gland. MATERIALS AND METHODS Formalin-fixed, paraffin-embedded tissue samples from patients pathologically diagnosed as lacrimal gland ACC (n=11) and salivary gland ACC (n=64) were used. Immunochemistry was used to measure expression of autophagy related proteins [beclin-1, light chain (LC) 3A, LC3B, p62, and BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3)] and ROS related proteins [catalase, thioredoxinreductase, glutathione S-transferasepi (GSTpi), thioredoxin interacting protein, and manganese superoxide dismutase (MnSOD)]. The prognostic factors related to disease-free and overall survival (OS) in lacrimal gland ACC by log-rank tests, were determined. RESULTS GSTpi in stromal cells was more highly expressed in lacrimal gland ACC (p=0.006), however, MnSOD in epithelial cells was expressed more in salivary gland ACC (p=0.046). LC3B positivity and BNIP3 positivity in epithelial component were associated with shorter disease-free survival (both p=0.002), and LC3A positivity in stromal component was the factor related to shorter OS (p=0.005). CONCLUSION This is the first study to demonstrate the expression of autophagy and ROS related proteins in lacrimal gland ACC in comparison with the salivary gland ACC, which would provide a basis for further study of autophagy and ROS mechanism as novel therapeutic targets in lacrimal gland ACC.
Collapse
Affiliation(s)
- Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Won Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Sook Yoon
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
44
|
Ma M, Zhao H, Zhao H, Binari R, Perrimon N, Li Z. Wildtype adult stem cells, unlike tumor cells, are resistant to cellular damages in Drosophila. Dev Biol 2016; 411:207-216. [PMID: 26845534 DOI: 10.1016/j.ydbio.2016.01.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/31/2016] [Accepted: 01/31/2016] [Indexed: 11/26/2022]
Abstract
Adult stem cells or residential progenitor cells are critical to maintain the structure and function of adult tissues (homeostasis) throughout the lifetime of an individual. Mis-regulation of stem cell proliferation and differentiation often leads to diseases including cancer, however, how wildtype adult stem cells and cancer cells respond to cellular damages remains unclear. We find that in the adult Drosophila midgut, intestinal stem cells (ISCs), unlike tumor intestinal cells, are resistant to various cellular damages. Tumor intestinal cells, unlike wildtype ISCs, are easily eliminated by apoptosis. Further, their proliferation is inhibited upon autophagy induction, and autophagy-mediated tumor inhibition is independent of caspase-dependent apoptosis. Interestingly, inhibition of tumorigenesis by autophagy is likely through the sequestration and degradation of mitochondria, as compromising mitochondria activity in these tumor models mimics the induction of autophagy and increasing the production of mitochondria alleviates the tumor-suppression capacity of autophagy. Together, these data demonstrate that wildtype adult stem cells and tumor cells show dramatic differences in sensitivity to cellular damages, thus providing potential therapeutic implications targeting tumorigenesis.
Collapse
Affiliation(s)
- Meifang Ma
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Hang Zhao
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Hanfei Zhao
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Richard Binari
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China.
| |
Collapse
|
45
|
Wang C, Li T, Tang S, Zhao D, Zhang C, Zhang S, Deng S, Zhou Y, Xiao X. Thapsigargin induces apoptosis when autophagy is inhibited in HepG2 cells and both processes are regulated by ROS-dependent pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 41:167-179. [PMID: 26708201 DOI: 10.1016/j.etap.2015.11.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 06/05/2023]
Abstract
Thapsigargin (TG), is widely used to induce endoplasmic reticular stress. Treated with TG for a long time, cells suffer the unfolded protein response (UPR) to elude apoptosis, but may activate autophagy. However, the switch between autophagy and apoptosis is unclear. To clarify the key signal for selection of these two protective responses, we studied the correlation of autophagy and apoptosis in HepG2 cells exposed to TG with time. TG induced apoptosis in HepG2 cells was evidenced by typical cell morphological changes and the activation of caspase-12, caspase-9 and caspase-3. Meanwhile, cytochrome c was released following with the dissipation of mitochondrial membrane potential (MMP), and the ratio of Bax/Bcl-2 was increased. TG-induced autophagy was confirmed by the accumulation of MDC, GFP-LC3 staining autophagic vacuoles, and the improved expression of LC3 II and Beclin-1. Additionally, inhibited autophagy via chloroquine (CQ) markedly enhanced the apoptosis induced by TG, which was linked to the Bcl-2 family. Furthermore, TG induced the generation of reactive oxygen species (ROS), and the ROS scavenger effectively suppressed TG-induced apoptosis and autophagy. All these results proved that restraint of autophagy may enhance TG-induced apoptosis through increasing the Bax/Bcl-2 ratio and both processes were regulated by ROS.
Collapse
Affiliation(s)
- Congcong Wang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China; Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, Ministry of Education, College of Marine Sciences, Shanghai Ocean University, Shanghai 201306, People's Republic of China
| | - Tao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, People's Republic of China
| | - Shusheng Tang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Dongxu Zhao
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Chaoming Zhang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Shen Zhang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Sijun Deng
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Yan Zhou
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Xilong Xiao
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China.
| |
Collapse
|
46
|
Zhou Y, Rucker EB, Zhou BP. Autophagy regulation in the development and treatment of breast cancer. Acta Biochim Biophys Sin (Shanghai) 2016; 48:60-74. [PMID: 26637829 DOI: 10.1093/abbs/gmv119] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/21/2015] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a major catabolic process in which intracellular membrane structures, protein complexes, and lysosomes are formed as lysoautophagosome to degrade and renew cytoplasmic components. Autophagy is physiologically a strategy and mechanism for cellular homeostasis as well as adaptation to stress, and thus alterations in the autophagy machinery may lead to diverse pathological conditions. The role of autophagy in cancer is complex, and the current literature reflects this as a 'double-edged sword'. Autophagy shows promise as a novel therapeutic target in various types of breast cancer, inhibiting or increasing treatment efficacy in a context- and cell-type-dependent manner. This review aims to summarize the recent advances in the understanding of the mechanisms by which key modulators of autophagy participate in cancer metastasis, highlight different autophagy-deficient murine models for breast cancer study, and provide further impetus for the modulation of autophagy in anticancer therapy.
Collapse
Affiliation(s)
- Yuting Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky School of Medicine, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| | - Edmund B Rucker
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky School of Medicine, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| |
Collapse
|
47
|
BARTSCH GEORG, JENNEWEIN LUKAS, HARTER PATRICKN, ANTONIETTI PATRICK, BLAHETA ROMANA, KVASNICKA HANSMICHAEL, KÖGEL DONAT, HAFERKAMP AXEL, MITTELBRONN MICHEL, MANI JENS. Autophagy-associated proteins BAG3 and p62 in testicular cancer. Oncol Rep 2015; 35:1629-35. [DOI: 10.3892/or.2015.4505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/03/2015] [Indexed: 11/06/2022] Open
|
48
|
Kuger S, Flentje M, Djuzenova CS. Simultaneous perturbation of the MAPK and the PI3K/mTOR pathways does not lead to increased radiosensitization. Radiat Oncol 2015; 10:214. [PMID: 26498922 PMCID: PMC4619315 DOI: 10.1186/s13014-015-0514-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/17/2015] [Indexed: 01/05/2023] Open
Abstract
Background The mitogen-activated protein kinases (MAPK) and the phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathways are intertwined on various levels and simultaneous inhibition reduces tumorsize and prolonges survival synergistically. Furthermore, inhibiting these pathways radiosensitized cancer cells in various studies. To assess, if phenotypic changes after perturbations of this signaling network depend on the genetic background, we integrated a time series of the signaling data with phenotypic data after simultaneous MAPK/ERK kinase (MEK) and PI3K/mTOR inhibition and ionizing radiation (IR). Methods The MEK inhibitor AZD6244 and the dual PI3K/mTOR inhibitor NVP-BEZ235 were tested in glioblastoma and lung carcinoma cells, which differ in their mutational status in the MAPK and the PI3K/mTOR pathways. Effects of AZD6244 and NVP-BEZ235 on the proliferation were assessed using an ATP assay. Drug treatment and IR effects on the signaling network were analyzed in a time-dependent manner along with measurements of phenotypic changes in the colony forming ability, apoptosis, autophagy or cell cycle. Results Both inhibitors reduced the tumor cell proliferation in a dose-dependent manner, with NVP-BEZ235 revealing the higher anti-proliferative potential. Our Western blot data indicated that AZD6244 and NVP-BEZ235 perturbed the MAPK and PI3K/mTOR signaling cascades, respectively. Additionally, we confirmed crosstalks and feedback loops in the pathways. As shown by colony forming assay, the AZD6244 moderately radiosensitized cancer cells, whereas NVP-BEZ235 caused a stronger radiosensitization. Combining both drugs did not enhance the NVP-BEZ235-mediated radiosensitization. Both inhibitors caused a cell cycle arrest in the G1-phase, whereas concomitant IR and treatment with the inhibitors resulted in cell line- and drug-specific cell cycle alterations. Furthermore, combining both inhibitors synergistically enhanced a G1-phase arrest in sham-irradiated glioblastoma cells and induced apoptosis and autophagy in both cell lines. Conclusion Perturbations of the MEK and the PI3K pathway radiosensitized tumor cells of different origins and the combination of AZD6244 and NVP-BEZ235 yielded cytostatic effects in several tumor entities. However, this is the first study assessing, if the combination of both drugs also results in synergistic effects in terms of radiosensitivity. Our study demonstrates that simultaneous treatment with both pathway inhibitors does not lead to synergistic radiosensitization but causes cell line-specific effects. Electronic supplementary material The online version of this article (doi:10.1186/s13014-015-0514-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Kuger
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany.
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Cholpon S Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Liang DH, El-Zein R, Dave B. Autophagy Inhibition to Increase Radiosensitization in Breast Cancer. ACTA ACUST UNITED AC 2015; 6. [PMID: 26613064 DOI: 10.4172/2155-9619.1000254] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Currently, many breast cancer patients with localized breast cancer undergo breast-conserving therapy, consisting of local excision followed by radiation therapy. Following radiation therapy, breast cancer cells are noted to undergo induction of autophagy, development of radioresistance, and enrichment of breast cancer stem cell subpopulations. It is hypothesized that inhibition of the cytoprotective autophagy that arises following radiation therapy increases radiosensitivity and confers longer relapse-free survival by eliminating tumor-initiating breast cancer stem cells. Therefore, we reviewed the controversial role of autophagy in breast cancer tumorigenesis and progression, autophagy induction by radiotherapy, and utilization of autophagy inhibitors to increase radiosensitivity of breast cancer and to target radioresistant breast cancer stem cells.
Collapse
Affiliation(s)
- Diana Hwang Liang
- Department of Surgery, Houston Methodist Hospital, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Randa El-Zein
- Department of Radiology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Bhuvanesh Dave
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
50
|
Jung YY, Lee YK, Koo JS. The potential of Beclin 1 as a therapeutic target for the treatment of breast cancer. Expert Opin Ther Targets 2015; 20:167-78. [PMID: 26357854 DOI: 10.1517/14728222.2016.1085971] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Beclin 1 plays a crucial role in autophagy via the Beclin 1 interactome, and is involved in various biological processes such as protein sorting, chemokinesis, and cell death. Via these biologic functions, Beclin 1 contributes to both tumor suppression and tumor progression. AREAS COVERED Beclin 1 plays a key biologic function on cell homeostasis and affects tumorigenesis. In this review, detailing up-to-date knowledge on the tumorigenic role of Beclin 1, its implication in breast cancer, and its utility as a breast cancer-specific drug target is discussed. EXPERT OPINION Because Beclin 1 is expressed in breast cancer cells, Beclin 1 could be a unique, effective drug target for the prevention and treatment of breast cancer. However, the expression of Beclin 1 varies according to cancer molecular subtypes, and Beclin 1 is involved in both breast cancer suppression and tumor progression; therefore, the decision of using a Beclin 1 inducer or inhibitor should be made based on breast cancer stage and subtype.
Collapse
Affiliation(s)
- Yoon Yang Jung
- a Yonsei University College of Medicine, Severance Hospital, Department of Pathology , 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, South Korea ;
| | - Yu Kyung Lee
- a Yonsei University College of Medicine, Severance Hospital, Department of Pathology , 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, South Korea ;
| | - Ja Seung Koo
- a Yonsei University College of Medicine, Severance Hospital, Department of Pathology , 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, South Korea ;
| |
Collapse
|