1
|
Koizume S, Miyagi Y. Adaptation mechanisms in cancer: Lipid metabolism under hypoxia and nutrient deprivation as a target for novel therapeutic strategies (Review). Mol Med Rep 2025; 31:83. [PMID: 39886950 PMCID: PMC11799873 DOI: 10.3892/mmr.2025.13448] [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: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025] Open
Abstract
Tumor tissues generally exist in a relatively hypovascular state, and cancer cells must adapt to severe tissue conditions with a limited molecular oxygen and nutrient supply for their survival. Lipid metabolism serves a role in this adaptation. Lipids are supplied not only through the bloodstream but also through autonomous synthesis by cancer cells, and they function as sources of adenosine triphosphate and cell components. Although cancer‑associated lipid metabolism has been widely reviewed, how this metabolism responds to the tumor environment with poor molecular oxygen and nutrient supply remains to be fully discussed. The main aim of the present review was to summarize the findings on this issue and to provide insights into how cancer cells adapt to better cope with metabolic stresses within tumors. It may be suggested that diverse types of lipid metabolism have a role in enabling cancer cells to adapt to both hypoxia and nutrient‑poor conditions. Gaining a deeper understanding of these molecular mechanisms may reveal novel possibilities of exploration for cancer treatment.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa 241-8515, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Kanagawa 241-8515, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa 241-8515, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Kanagawa 241-8515, Japan
| |
Collapse
|
2
|
Li M, Feng J, Zhao K, Huang T, Zhang B, Yang Y, Sun A, Lin Q, Shao G. LSD1 Demethylates and Destabilizes Autophagy Protein LC3B in Ovarian Cancer. Biomolecules 2024; 14:1377. [PMID: 39595554 PMCID: PMC11591952 DOI: 10.3390/biom14111377] [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: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy is a complex cellular process that can either promote or inhibit cancer progression and development, depending on the context and molecular regulation involved. This study investigates how LSD1 regulates autophagy in ovarian cancer by interacting with the autophagy protein LC3B. Utilizing the bioinformatic analysis of TCGA, CPTAC, and GEO datasets, as well as immunohistochemistry in ovarian cancer patients, we explored the expression association between LSD1 and LC3B. Molecular mechanisms were further analyzed using Western blotting, immunoprecipitation, and GST pull-down assays. Our findings reveal that LSD1 binds to LC3B via its SWIRM domain, and high levels of LSD1 are closely associated with aggressive ovarian cancer and poor patient outcomes. Mechanistically, LSD1 demethylates LC3B, leading to decreased LC3B stability. The observed inverse correlation between LSD1 expression and LC3B protein levels in clinical samples underscores the need for further investigation to elucidate how reduced LC3B protein levels induced by LSD1 demethylation may contribute to ovarian cancer.
Collapse
Affiliation(s)
- Mingyang Li
- Institute of Urinary System Diseases, The Affiliated People’s Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang 212002, China;
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jie Feng
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Kangrong Zhao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Bowen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yifan Yang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Aiqin Sun
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Genbao Shao
- Institute of Urinary System Diseases, The Affiliated People’s Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang 212002, China;
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
3
|
Li D, Geng D, Wang M. Advances in natural products modulating autophagy influenced by cellular stress conditions and their anticancer roles in the treatment of ovarian cancer. FASEB J 2024; 38:e70075. [PMID: 39382031 DOI: 10.1096/fj.202401409r] [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: 06/20/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024]
Abstract
Autophagy is a conservative catabolic process that typically serves a cell-protective function. Under stress conditions, when the cellular environment becomes unstable, autophagy is activated as an adaptive response for self-protection. Autophagy delivers damaged cellular components to lysosomes for degradation and recycling, thereby providing essential nutrients for cell survival. However, this function of promoting cell survival under stress conditions often leads to malignant progression and chemotherapy resistance in cancer. Consequently, autophagy is considered a potential target for cancer therapy. Herein, we aim to review how natural products act as key modulators of autophagy by regulating cellular stress conditions. We revisit various stressors, including starvation, hypoxia, endoplasmic reticulum stress, and oxidative stress, and their regulatory relationship with autophagy, focusing on recent advances in ovarian cancer research. Additionally, we explore how polyphenolic compounds, flavonoids, alkaloids, terpenoids, and other natural products modulate autophagy mediated by stress responses, affecting the malignant biological behavior of cancer. Furthermore, we discuss their roles in ovarian cancer therapy. This review emphasizes the importance of natural products as valuable resources in cancer therapeutics, highlighting the need for further exploration of their potential in regulating autophagy. Moreover, it provides novel insights and potential therapeutic strategies in ovarian cancer by utilizing natural products to modulate autophagy.
Collapse
Affiliation(s)
- Dongxiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Danbo Geng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Cai J, Hu Q, He Z, Chen X, Wang J, Yin X, Ma X, Zeng J. Scutellaria baicalensis Georgi and Their Natural Flavonoid Compounds in the Treatment of Ovarian Cancer: A Review. Molecules 2023; 28:5082. [PMID: 37446743 DOI: 10.3390/molecules28135082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common types of cancer in women with a high mortality rate, and the treatment of OC is prone to high recurrence rates and side effects. Scutellaria baicalensis (SB) is a herbal medicine with good anti-cancer activity, and several studies have shown that SB and its flavonoids have some anti-OC properties. This paper elucidated the common pathogenesis of OC, including cell proliferation and cell cycle regulation, cell invasion and metastasis, apoptosis and autophagy, drug resistance and angiogenesis. The mechanisms of SB and its flavonoids, wogonin, baicalein, baicalin, Oroxylin A, and scutellarein, in the treatment of OC, are revealed, such as wogonin inhibits proliferation, induces apoptosis, inhibits invasion and metastasis, and increases the cytotoxicity of the drug. Baicalein also inhibits vascular endothelial growth factor (VEGF) expression etc. Analyzing their advantages and disadvantages in treating OC provides a new perspective on the role of SB and its flavonoids in OC treatment. It serves as a resource for future OC research and development.
Collapse
Affiliation(s)
- Jiaying Cai
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhelin He
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiaoyan Chen
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Jian Wang
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiang Yin
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
5
|
Abstract
Autophagy is a self-digestion process by which misfolded proteins and damaged organelles in eukaryotic cells are degraded to maintain cellular homeostasis. This process is involved in the tumorigenesis, metastasis, and chemoresistance of various tumors such as ovarian cancer (OC). Noncoding RNAs (ncRNAs), mainly including microRNAs, long noncoding RNAs, and circular RNAs, have been extensively investigated in cancer research for their roles in the regulation of autophagy. Recent studies have shown that in OC cells, ncRNAs can modulate the formation of autophagosomes, which affect tumor progression and chemoresistance. An understanding of the role of autophagy in OC progression, treatment, and prognosis is important, and the identification of the regulatory roles of ncRNAs in autophagy leads to intervention strategies for OC therapy. This review summarizes the role of autophagy in OC and discusses the role of ncRNA-mediated autophagy in OC, as an understanding of these roles may contribute to the development of potential therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Cong Feng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, P.R. China
- Heilongjiang University of Chinese Medicine, Harbin 150040, P.R. China
| | - Xingxing Yuan
- Heilongjiang University of Chinese Medicine, Harbin 150040, P.R. China
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, P.R. China
| |
Collapse
|
6
|
Guo N, Yang A, Farooq FB, Kalaria S, Moss E, DeVorkin L, Lesperance M, Bénard F, Wilson D, Tinker AV, Nathoo FS, Hamilton PT, Lum JJ. CD8 + T cell infiltration is associated with improved survival and negatively correlates with hypoxia in clear cell ovarian cancer. Sci Rep 2023; 13:6530. [PMID: 37085560 PMCID: PMC10121667 DOI: 10.1038/s41598-023-30655-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/27/2023] [Indexed: 04/23/2023] Open
Abstract
Unlike other histological types of epithelial ovarian carcinoma, clear cell ovarian carcinoma (CCOC) has poor response to therapy. In many other carcinomas, expression of the hypoxia-related enzyme Carbonic anhydrase IX (CAIX) by cancer cells is associated with poor prognosis, while the presence of CD8 + tumor-infiltrating lymphocytes (TIL) is positively prognostic. We employed [18F]EF5-PET/CT imaging, transcriptome profiling, and spatially-resolved histological analysis to evaluate relationships between CAIX, CD8, and survival in CCOC. Tissue microarrays (TMAs) were evaluated for 218 cases in the Canadian COEUR study. Non-spatial relationships between CAIX and CD8 were investigated using Spearman rank correlation, negative binomial regression and gene set enrichment analysis. Spatial relationships at the cell level were investigated using the cross K-function. Survival analysis was used to assess the relationship of CAIX and CD8 with patient survival for 154 cases. CD8 + T cell infiltration positively predicted survival with estimated hazard ratio 0.974 (95% CI 0.950, 1000). The negative binomial regression analysis found a strong TMA effect (p-value < 0.0001). It also indicated a negative association between CD8 and CAIX overall (p-value = 0.0171) and in stroma (p-value = 0.0050) but not in tumor (p-value = 0.173). Examination of the spatial association between the locations of CD8 + T cells and CAIX cells found a significant amount of heterogeneity in the first TMA, while in the second TMA there was a clear signal indicating negative spatial association in stromal regions. These results suggest that hypoxia may contribute to immune exclusion, primarily mediated by effects in stroma.
Collapse
Affiliation(s)
- Nancy Guo
- Department of Mathematics and Statistics, University of Victoria, STN CSC, PO BOX 1700, Victoria, BC, V8W 2Y2, Canada
| | - Aijun Yang
- Department of Mathematics and Statistics, University of Victoria, STN CSC, PO BOX 1700, Victoria, BC, V8W 2Y2, Canada
| | | | - Shreena Kalaria
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, 2410 Lee Avenue, 3rd Floor, Victoria, BC, V8R 6V5, Canada
| | - Elena Moss
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, 2410 Lee Avenue, 3rd Floor, Victoria, BC, V8R 6V5, Canada
| | - Lindsay DeVorkin
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, 2410 Lee Avenue, 3rd Floor, Victoria, BC, V8R 6V5, Canada
| | - Mary Lesperance
- Department of Mathematics and Statistics, University of Victoria, STN CSC, PO BOX 1700, Victoria, BC, V8W 2Y2, Canada
| | - François Bénard
- Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Don Wilson
- Functional Imaging, BC Cancer Vancouver, Vancouver, Canada
| | - Anna V Tinker
- Medical Oncology, BC Cancer Vancouver, Vancouver, Canada
| | - Farouk S Nathoo
- Department of Mathematics and Statistics, University of Victoria, STN CSC, PO BOX 1700, Victoria, BC, V8W 2Y2, Canada.
| | - Phineas T Hamilton
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, 2410 Lee Avenue, 3rd Floor, Victoria, BC, V8R 6V5, Canada.
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, 2410 Lee Avenue, 3rd Floor, Victoria, BC, V8R 6V5, Canada.
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada.
| |
Collapse
|
7
|
Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S, Mansouri K. Autophagy: A challengeable paradox in cancer treatment. Cancer Med 2023. [PMID: 36760166 DOI: 10.1002/cam4.5577] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Autophagy is an intracellular degradation pathway conserved in all eukaryotes from yeast to humans. This process plays a quality-control role by destroying harmful cellular components under normal conditions, maintaining cell survival, and establishing cellular adaptation under stressful conditions. Hence, there are various studies indicating dysfunctional autophagy as a factor involved in the development and progression of various human diseases, including cancer. In addition, the importance of autophagy in the development of cancer has been highlighted by paradoxical roles, as a cytoprotective and cytotoxic mechanism. Despite extensive research in the field of cancer, there are many questions and challenges about the roles and effects suggested for autophagy in cancer treatment. The aim of this study was to provide an overview of the paradoxical roles of autophagy in different tumors and related cancer treatment options. METHODS In this study, to find articles, a search was made in PubMed and Google scholar databases with the keywords Autophagy, Autophagy in Cancer Management, and Drug Design. RESULTS According to the investigation, some studies suggest that several advanced cancers are dependent on autophagy for cell survival, so when cancer cells are exposed to therapy, autophagy is induced and suppresses the anti-cancer effects of therapeutic agents and also results in cell resistance. However, enhanced autophagy from using anti-cancer drugs causes autophagy-mediated cell death in several cancers. Because autophagy also plays roles in both tumor suppression and promotion further research is needed to determine the precise mechanism of this process in cancer treatment. CONCLUSION We concluded in this article, autophagy manipulation may either promote or hinder the growth and development of cancer according to the origin of the cancer cells, the type of cancer, and the behavior of the cancer cells exposed to treatment. Thus, before starting treatment it is necessary to determine the basal levels of autophagy in various cancers.
Collapse
Affiliation(s)
- Farnaz Ahmadi-Dehlaghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Biology, Payame Noor University, Tehran, Iran
| | - Parisa Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elahe Valipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sarah Kiani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Koizume S, Takahashi T, Nakamura Y, Yoshihara M, Ota Y, Sato S, Tadokoro H, Yokose T, Kato H, Miyagi E, Miyagi Y. Lipophagy-ICAM-1 pathway associated with fatty acid and oxygen deficiencies is involved in poor prognoses of ovarian clear cell carcinoma. Br J Cancer 2022; 127:462-473. [PMID: 35449452 PMCID: PMC9346109 DOI: 10.1038/s41416-022-01808-4] [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: 09/15/2021] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Serum starvation and hypoxia (SSH) mimics a stress condition in tumours. We have shown that intercellular adhesion molecule-1 (ICAM-1) protein is synergistically expressed in ovarian clear cell carcinoma (CCC) cells under SSH in response to an insufficient supply of fatty acids (FAs). This ICAM-1 expression is responsible for resistance against the lethal condition, thereby promoting tumour growth. However, the underlying mechanisms that link SSH-driven ICAM1 gene expression to impaired FA supply and its clinical relevance are unclear. METHODS The underlying mechanisms of how FA deficiency induces ICAM-1 expression in cooperation with hypoxia were analysed in vitro and in vivo. Clinical significance of CCC cell-derived ICAM-1 and the mechanism associated with the transcriptional synergism were also investigated. RESULTS ICAM-1 expression was mediated through lipophagy-driven lipid droplet degradation, followed by impaired FA-lipid droplet flow. Lipophagy induced ICAM1 expression through stabilisation of NFκB binding to the promoter region via Sam68 and hTERT. Analyses of clinical specimens revealed that expression of ICAM-1 and LC3B, an autophagy marker associated with lipophagy, significantly correlated with poor prognoses of CCC. CONCLUSIONS The lipophagy-ICAM-1 pathway induced under a tumour-like stress conditions contributes to CCC progression and is a potential therapeutic target for this aggressive cancer type.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515, Japan. .,Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, 241-8515, Japan.
| | - Tomoko Takahashi
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan
| | - Yoshiyasu Nakamura
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan
| | - Mitsuyo Yoshihara
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan
| | - Yukihide Ota
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan ,grid.268441.d0000 0001 1033 6139Department of Obstetrics, Gynecology, and Molecular Reproductive Science, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Shinya Sato
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan ,grid.414944.80000 0004 0629 2905Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, 241-8515 Japan
| | - Hiroko Tadokoro
- grid.414944.80000 0004 0629 2905Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515 Japan
| | - Tomoyuki Yokose
- grid.414944.80000 0004 0629 2905Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, 241-8515 Japan
| | - Hisamori Kato
- grid.414944.80000 0004 0629 2905Department of Gynecology, Kanagawa Cancer Center Hospital, Yokohama, 241-8515 Japan
| | - Etsuko Miyagi
- grid.268441.d0000 0001 1033 6139Department of Obstetrics, Gynecology, and Molecular Reproductive Science, Yokohama City University, Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, 241-8515, Japan. .,Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, 241-8515, Japan.
| |
Collapse
|
9
|
Kobayashi H, Imanaka S. Toward an understanding of tissue factor pathway inhibitor-2 as a novel serodiagnostic marker for clear cell carcinoma of the ovary. J Obstet Gynaecol Res 2021; 47:2978-2989. [PMID: 34184357 DOI: 10.1111/jog.14916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 12/19/2022]
Abstract
AIMS Tissue factor pathway inhibitor (TFPI)-2 has recently emerged as a serodiagnostic marker for patients with epithelial ovarian cancer (EOC), especially clear cell carcinoma (CCC). This review discusses the biological properties of TFPI-2 and why serum levels are elevated in CCC patients. METHODS A comprehensive literature search was conducted in PubMed up until March, 2021. RESULTS TFPI-2 is a Kunitz-type protease inhibitor and negatively regulates the enzymatic activities, such as plasmin. TFPI-2 has been characterized as a tumor suppressor gene and was frequently downregulated through promoter hypermethylation in various human cancers. In contrast, TFPI-2 was overexpressed only in CCC. TFPI-2 may be involved in the pathophysiology of CCC, possibly through regulation of coagulation system, stabilization of extracellular matrix (ECM), and induction of intracellular signal transduction. TFPI-2 suppresses tissue factor-induced hypercoagulation in a hypoxic environment. TFPI-2, secreted by CCC cells, platelets, and adjacent vascular endothelial cells, may suppress tumor growth and invasion through ECM remodeling. Nuclear TFPI-2 may suppress matrix metalloproteinase production via transcription factors and modulate caspase-mediated cell apoptosis. CCC cells may upregulate the TFPI-2 expression to adapt to survival in the demanding environment. TFPI-2 is secreted by CCC cells and enters the systemic circulation, resulting in elevated blood levels. DISCUSSION Serum TFPI-2 reflects the overexpression of TFPI-2 in CCC tissues and is a potential serodiagnostic marker. Further research is needed to explore the expression, clinical significance, biological function, and potential mechanism of TFPI-2 in CCC.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara, Japan.,Ms. Clinic MayOne, Kashihara, Nara, Japan
| | - Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara, Japan.,Ms. Clinic MayOne, Kashihara, Nara, Japan
| |
Collapse
|
10
|
Chen X, Sun Y, Wang B, Wang H. Prognostic significance of autophagy-related genes Beclin1 and LC3 in ovarian cancer: a meta-analysis. J Int Med Res 2021; 48:300060520968299. [PMID: 33238786 PMCID: PMC7705295 DOI: 10.1177/0300060520968299] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Beclin1 plays a central role in the activation of the autophagy signaling pathway. Beclin1 and LC3-related proteins are involved in the initial steps of autophagy, which are closely related to the occurrence and development of tumors. The current meta-analysis aimed to clarify the correlation between expression of Beclin1 and LC3 and prognosis of ovarian cancer. METHODS We searched PubMed, Embase, The Cochrane Library, Web of Science, and CNKI using predefined selection criteria. Pooled hazard ratios and relative risks with 95% confidence intervals were used to evaluate the correlation between autophagy-related genes Beclin1 and LC3 and overall survival (OS), progression-free survival (PFS), and International Federation of Gynecology and Obstetrics (FIGO) stage. RESULTS In total, 1497 patients from 10 articles were enrolled in this meta-analysis. Expression of Beclin1 was significantly correlated with improved OS and PFS, and increased expression of Beclin1 was correlated with early FIGO stage, but not with lymph node metastasis or histological grade. No association was found between LC3 expression and prognosis in patients with ovarian cancer. CONCLUSIONS Expression of Beclin1 is an independent risk factor for the progression of ovarian cancer. Thus, Beclin1 is a promising indicator in predicting prognosis in patients with ovarian cancer.
Collapse
Affiliation(s)
- Xinbei Chen
- Department of Gynecology, Fujian Cancer Hospital, Affiliated Cancer Hospital of 74551Fujian Medical University, Fuzhou, Fujian, China.,Department of Gynecology, Shengli Clinical Medical College of 74551Fujian Medical University, Fuzhou, Fujian, China
| | - Yang Sun
- Department of Gynecology, Fujian Cancer Hospital, Affiliated Cancer Hospital of 74551Fujian Medical University, Fuzhou, Fujian, China
| | - Bingrong Wang
- Department of Gynecology, Fujian Cancer Hospital, Affiliated Cancer Hospital of 74551Fujian Medical University, Fuzhou, Fujian, China.,Department of Gynecology, Shengli Clinical Medical College of 74551Fujian Medical University, Fuzhou, Fujian, China
| | - Huihui Wang
- Department of Gynecology, Fujian Cancer Hospital, Affiliated Cancer Hospital of 74551Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
11
|
LC3A positive "stone like structures" are differentially associated with survival outcomes and CD68 macrophage infiltration in patients with lung adenocarcinoma and squamous cell carcinoma. Lung Cancer 2021; 156:129-135. [PMID: 33962766 DOI: 10.1016/j.lungcan.2021.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/22/2022]
Abstract
AIMS The aim of the study was to analyse the prognostic and predictive value of LC3A positive' 'Stone Like Structures'' (SLSs) in a large cohort of patients with non-small cell lung carcinoma (NSCLC) and to check its relationship with tumor infiltrating lymphocytes (TILs) and PD-L1 expression. METHODS Tissue microarrays from 1015 patients diagnosed at the Institute of Pathology and Molecular Pathology, University Hospital Zurich, Switzerland, were stained for LC3A, PD-L1, CD3 and CD68 using automated tissue stainer Ventana Benchmark Ultra (Roche). TILs were assessed in matched haematoxylin and eosin stained slides. RESULTS LC3A positive SLSs, were significantly associated with worse overall (OS) and disease-free survival (DFS) outcomes in patients with lung adenocarcinoma (LADC) (HR = 2.4, 95 %CI(.994-1.008, p = 0.029) and HR = 3.9, 95 %CI (1.002-1.014), p = 0.002 respectively), whilst it was associated with better OS and DFS in patients with lung squamous cell carcinoma (LUSC), with marginal significance (HR = .99, 95 %CI(.975-1.011),p = 0.042 and HR = .99, 95 %CI (.975-1.008), p = 0.026). Multivariate analysis showed that LC3A SLSs are independent poor prognostic factor only in patients with LADC. In addition, LC3A SLSs, were negatively associated with CD68 count in LADC, whilst there was a positive correlation in LSCC. CONCLUSIONS LC3A SLSs are differentially associated with the survival outcomes and CD68 count in LADC and LSCC. Further studies are justified for the understanding the underlying biological mechanisms of this phenomenon.
Collapse
|
12
|
Thongchot S, Jamjuntra P, Therasakvichya S, Warnnissorn M, Ferraresi A, Thuwajit P, Isidoro C, Thuwajit C. Interleukin‑8 released by cancer‑associated fibroblasts attenuates the autophagy and promotes the migration of ovarian cancer cells. Int J Oncol 2021; 58:14. [PMID: 33649784 PMCID: PMC7949624 DOI: 10.3892/ijo.2021.5194] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment composed of a mixture of stromal cells and their secretions has a marked impact on cancer progression. In particular, soluble factors and metabolites contribute to malignancy through the dysregulation of autophagy in cancer cells. The present study investigated the effects of ovarian cancer‑associated fibroblasts (OVCAFs) with their secretory substances on the autophagy and migration of ovarian cancer cells. The conditioned‑medium (CM) of OVCAFs isolated from fresh human ovarian cancer tissues was analyzed for the levels of 27 common cytokines/chemokines using a cytokine array. Autophagy in cancer cells was assessed by determining the expression of the vacuolar form of LC3 by western blot analysis and immunofluorescence. Cancer cell migration was assessed by Transwell migration assay. Interleukin (IL)‑8 was found to be the most highly upregulated cytokine among the cytokines/chemokines found in the OVCAF‑CM. The role of IL‑8 in ovarian cancer cell migration and its mechanistic link with autophagy was investigated. Recombinant human IL‑8 (rhIL‑8) stimulated the migration of SKOV3 and Kuramochi ovarian cancer cells, and concurrently downregulated basal autophagy, in concentration‑dependent manner. Compared to the CM of control counterpart normal fibroblasts isolated from benign ovaries (OVNF‑CM), the CM from 3 OVCAF isolates (namely, OVCAF‑9, ‑20 and ‑43) exerted effects similar to rhIL‑8 on both cancer cell lines. The pharmacological induction of autophagy with rapamycin or metformin attenuated the pro‑migratory effects of IL‑8. Neutralizing anti‑IL‑8 antibody counteracted the inhibitory effect of OVCAF‑CM on basal autophagy. On the whole, the present study highlights the involvement of IL‑8 released by CAFs in the ovarian tumor microenvironment in promoting cancer cell migration through the suppression of autophagy.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pranisa Jamjuntra
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suwanit Therasakvichya
- Department of Gynecology and Obstetrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Malee Warnnissorn
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, University of Eastern Piedmont 'A. Avogadro', I‑28100 Novara, Italy
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, University of Eastern Piedmont 'A. Avogadro', I‑28100 Novara, Italy
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
13
|
Tang Y, Zhang Y, Liu S, Sun Z, Wang C, Li L, Zhou W, Cheng S. 14-3-3ζ binds to and stabilizes phospho-beclin 1 S295 and induces autophagy in hepatocellular carcinoma cells. J Cell Mol Med 2019; 24:954-964. [PMID: 31709727 PMCID: PMC6933394 DOI: 10.1111/jcmm.14806] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/21/2019] [Accepted: 10/20/2019] [Indexed: 12/21/2022] Open
Abstract
Data from The Cancer Genome Atlas (TCGA) indicate that the expression levels of 14‐3‐3ζ and beclin 1 (a key molecule involved in cellular autophagy) are up‐regulated and positively correlated with each other (R = .5, P < .05) in HCC tissues. Chemoresistance developed in hepatoma cancer cells is associated with autophagy initiation. This study aimed to explore 14‐3‐3ζ’s role in regulating autophagy in HCC cells, with a focus on beclin 1. The co‐localization of 14‐3‐3ζ and beclin 1 was detectable in primary HCC tissues. To simulate in vivo tumour microenvironment (hypoxia), CSQT‐2 and HCC‐LM3 cells were exposed to 2% oxygen for 24 hours. The protein levels of 14‐3‐3ζ and phospho‐beclin 1S295 peaked at 12 hours following hypoxia. Meanwhile, the strongest autophagy flux occurred: LC3II was increased, and p62 was decreased significantly. By sequencing the coding area of BECN 1 gene of CSQT‐2 and HCC‐LM3 cells, we found that the predicted translational products of BECN 1 gene contained RLPS295VP (R, arginine; L, leucine; P, proline; S, serine; V, valine), a classic 14‐3‐3ζ binding motif. CO‐IP results confirmed that 14‐3‐3ζ bound to beclin 1, and this connection was markedly weakened when S295 was mutated into A295 (alanine). Further, 14‐3‐3ζ overexpression prevented phospho‐beclin 1S295 from degradation and enhanced its binding to VPS34, whilst its knockdown accelerated the degradation. Additionally, 14‐3‐3ζ enhanced the chemoresistance of HCC cells to cis‐diammined dichloridoplatium by activating autophagy. Our work reveals that 14‐3‐3ζ binds to and stabilizes phospho‐beclin 1S295 and induces autophagy in HCC cells to resist chemotherapy.
Collapse
Affiliation(s)
- Yufu Tang
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Yibing Zhang
- Department of Medical Affairs, The General Hospital of Northern Theater Command, Shenyang, China
| | - Shupeng Liu
- Department of Gynecology and Obstetrics, Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhongyi Sun
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Chunhui Wang
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Longfei Li
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Wenping Zhou
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Shuqun Cheng
- Department of Liver Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
14
|
Van Nest SJ, Nicholson LM, Pavey N, Hindi MN, Brolo AG, Jirasek A, Lum JJ. Raman spectroscopy detects metabolic signatures of radiation response and hypoxic fluctuations in non-small cell lung cancer. BMC Cancer 2019; 19:474. [PMID: 31109312 PMCID: PMC6528330 DOI: 10.1186/s12885-019-5686-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/08/2019] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Radiation therapy is a standard form of treating non-small cell lung cancer, however, local recurrence is a major issue with this type of treatment. A better understanding of the metabolic response to radiation therapy may provide insight into improved approaches for local tumour control. Cyclic hypoxia is a well-established determinant that influences radiation response, though its impact on other metabolic pathways that control radiosensitivity remains unclear. METHODS We used an established Raman spectroscopic (RS) technique in combination with immunofluorescence staining to measure radiation-induced metabolic responses in human non-small cell lung cancer (NSCLC) tumour xenografts. Tumours were established in NOD.CB17-Prkdcscid/J mice, and were exposed to radiation doses of 15 Gy or left untreated. Tumours were harvested at 2 h, 1, 3 and 10 days post irradiation. RESULTS We report that xenografted NSCLC tumours demonstrate rapid and stable metabolic changes, following exposure to 15 Gy radiation doses, which can be measured by RS and are dictated by the extent of local tissue oxygenation. In particular, fluctuations in tissue glycogen content were observed as early as 2 h and as late as 10 days post irradiation. Metabolically, this signature was correlated to the extent of tumour regression. Immunofluorescence staining for γ-H2AX, pimonidazole and carbonic anhydrase IX (CAIX) correlated with RS-identified metabolic changes in hypoxia and reoxygenation following radiation exposure. CONCLUSION Our results indicate that RS can identify sequential changes in hypoxia and tumour reoxygenation in NSCLC, that play crucial roles in radiosensitivity.
Collapse
Affiliation(s)
- Samantha J. Van Nest
- Department of Physics and Astronomy, University of Victoria, PO BOX 1700 STN CSC, Victoria, BC V8W 2Y2 Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, 2410 Lee Avenue, Victoria, BC V8R 6V5 Canada
| | - Leah M. Nicholson
- Trev and Joyce Deeley Research Centre, BC Cancer, 2410 Lee Avenue, Victoria, BC V8R 6V5 Canada
| | - Nils Pavey
- Trev and Joyce Deeley Research Centre, BC Cancer, 2410 Lee Avenue, Victoria, BC V8R 6V5 Canada
| | - Mathew N. Hindi
- Trev and Joyce Deeley Research Centre, BC Cancer, 2410 Lee Avenue, Victoria, BC V8R 6V5 Canada
| | - Alexandre G. Brolo
- Department of Chemistry, University of Victoria, PO BOX 3065, Victoria, BC V8W 3V6 Canada
| | - Andrew Jirasek
- Department of Physics, I.K. Barber School of Arts and Sciences, University of British Columbia-Okanagan, 3187 University Way, Kelowna, BC V1V 1V7 Canada
| | - Julian J. Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, 2410 Lee Avenue, Victoria, BC V8R 6V5 Canada
- Department of Biochemistry and Microbiology, University of Victoria, PO BOX 1700 STN CSC, Victoria, BC V8W 2Y2 Canada
| |
Collapse
|
15
|
Sivridis E, Koukourakis IM, Arelaki S, Balaska K, Karpouzis A, Giatromanolaki A. Patterns of LC3A Autophagy Protein Expression in Keratoacanthomas. Head Neck Pathol 2019; 14:150-155. [PMID: 30977096 PMCID: PMC7021871 DOI: 10.1007/s12105-019-01033-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/18/2019] [Indexed: 11/29/2022]
Abstract
To investigate the expression patterns of autophagy marker light chain protein 3 (LC3A) in keratoacanthoma (KA). KAs are generally regarded as benign but malignant behavior, including rare metastases, may occur. 85 KAs were assessed for the LC3A autophagic protein by immunohistochemistry. Diffuse cytoplasmic staining and a "stone-like structure" (SLS) characterized positive expression. Thirty-four out of 85 KAs (40%) had diffuse cytoplasmic LC3A immunostaining (percentage of positive cells ranging from 5 to 60%). In contrast, only 4 of the 85 KAs (4.7%) expressed SLSs. Only one SLS was detected per histologic section of each tumor. The p53 oncoprotein was encountered in all cases with expression ranging from 1 to 90% of cells (median 30%). The Ki-67 index was expressed in 63 cases (74% of cases; range 1-50% of cells; median value 5%). Neither of these two parameters nor diffuse cytoplasmic LC3A staining was significantly correlated with SLS expression or lack thereof. Expression of SLSs, a hallmark of malignancy, was found in 4.7% of KAs. Further study is necessary to determine whether this fraction represents the exceptional cases that harbor latent malignant potential.
Collapse
Affiliation(s)
- Efthimios Sivridis
- Departments of Pathology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Ioannis M. Koukourakis
- Departments of Pathology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Stella Arelaki
- Departments of Pathology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Kostantina Balaska
- Departments of Pathology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Antonios Karpouzis
- Departments of Dermatology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| | - Alexandra Giatromanolaki
- Departments of Pathology, Democritus University of Thrace Medical School and University General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| |
Collapse
|
16
|
Che J, Wang W, Huang Y, Zhang L, Zhao J, Zhang P, Yuan X. miR-20a inhibits hypoxia-induced autophagy by targeting ATG5/FIP200 in colorectal cancer. Mol Carcinog 2019; 58:1234-1247. [PMID: 30883936 DOI: 10.1002/mc.23006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 01/07/2023]
Abstract
Autophagy is a highly conserved lysosome-mediated protective cellular process in which cytosolic components, including damaged organelles and long-lived proteins, are cleared. Many studies have shown that autophagy was upregulated in hypoxic regions. However, the precise molecular mechanism of hypoxia-induced autophagy in colorectal cancer (CRC) is still elusive. In this study, we found that miR-20a was significantly downregulated under hypoxia in colon cancer cells, and overexpression of miR-20a alleviated hypoxia-induced autophagy. Moreover, miR-20a inhibits the hypoxia-induced autophagic flux by targeting multiple key regulators of autophagy, including ATG5 and FIP200. Furthermore, by dual-luciferase assay we demonstrated that miR-20a directly targeted the 3'-untranslated region of ATG5 and FIP200, regulating their messenger RNA and protein levels. In addition, reintroduction of exogenous ATG5 or FIP200 partially reversed miR-20a-mediated autophagy inhibition under hypoxia. A negative correlation between miR-20a and its target genes is observed in the hypoxic region of colon cancer tissues. Taken together, our findings suggest that hypoxia-mediated autophagy was regulated by miR-20a/ATG5/FI200 signaling pathway in CRC. miR-20a-mediated autophagy defect that might play an important role in hypoxia-induced autophagy during colorectal tumorigenesis.
Collapse
Affiliation(s)
- Jing Che
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Demonstration Center for Experimental Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Wenshan Wang
- Department of Cell and Developmental Biology, Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Young AN, Herrera D, Huntsman AC, Korkmaz MA, Lantvit DD, Mazumder S, Kolli S, Coss CC, King S, Wang H, Swanson SM, Kinghorn AD, Zhang X, Phelps MA, Aldrich LN, Fuchs JR, Burdette JE. Phyllanthusmin Derivatives Induce Apoptosis and Reduce Tumor Burden in High-Grade Serous Ovarian Cancer by Late-Stage Autophagy Inhibition. Mol Cancer Ther 2018; 17:2123-2135. [PMID: 30018048 PMCID: PMC6168422 DOI: 10.1158/1535-7163.mct-17-1195] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/06/2018] [Accepted: 07/13/2018] [Indexed: 11/16/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) is a lethal gynecological malignancy with a need for new therapeutics. Many of the most widely used chemotherapeutic drugs are derived from natural products or their semi-synthetic derivatives. We have developed potent synthetic analogues of a class of compounds known as phyllanthusmins, inspired by natural products isolated from Phyllanthus poilanei Beille. The most potent analogue, PHY34, had the highest potency in HGSOC cell lines in vitro and displayed cytotoxic activity through activation of apoptosis. PHY34 exerts its cytotoxic effects by inhibiting autophagy at a late stage in the pathway, involving the disruption of lysosomal function. The autophagy activator, rapamycin, combined with PHY34 eliminated apoptosis, suggesting that autophagy inhibition may be required for apoptosis. PHY34 was readily bioavailable through intraperitoneal administration in vivo where it significantly inhibited the growth of cancer cell lines in hollow fibers, as well as reduced tumor burden in a xenograft model. We demonstrate that PHY34 acts as a late-stage autophagy inhibitor with nanomolar potency and significant antitumor efficacy as a single agent against HGSOC in vivo This class of compounds holds promise as a potential, novel chemotherapeutic and demonstrates the effectiveness of targeting the autophagic pathway as a viable strategy for combating ovarian cancer. Mol Cancer Ther; 17(10); 2123-35. ©2018 AACR.
Collapse
Affiliation(s)
- Alexandria N Young
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Denisse Herrera
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Andrew C Huntsman
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Melissa A Korkmaz
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois
| | - Daniel D Lantvit
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Sarmistha Mazumder
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shamalatha Kolli
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Christopher C Coss
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Salane King
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Hongyan Wang
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Steven M Swanson
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xiaoli Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Mitch A Phelps
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Leslie N Aldrich
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Joanna E Burdette
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
18
|
Chan KK, Wong OGW, Wong ESY, Chan KKL, Ip PPC, Tse KY, Cheung ANY. Impact of iASPP on chemoresistance through PLK1 and autophagy in ovarian clear cell carcinoma. Int J Cancer 2018; 143:1456-1469. [PMID: 29663364 DOI: 10.1002/ijc.31535] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/15/2018] [Accepted: 03/13/2018] [Indexed: 12/17/2023]
Abstract
Ovarian clear cell carcinoma (OCCC) is a type of epithelial ovarian cancer that is strongly associated with endometriosis, resistance against conventional chemotherapy and thus poorer prognosis. The expression of inhibitory member of the ASPP family proteins (iASPP) and Polo-like kinase (PLK)1 were significantly higher in OCCC compared to benign cystadenomas and endometriosis. Both protein expressions were found to correlate with chemoresistance in patients with OCCC while high iASPP expression alone was significantly associated with a poor patient survival. The growth of OCCC cell lines, OVTOKO and KK, were inhibited after iASPP silencing. Such effect was related to senescence triggering as evidenced by increased SA-β-Gal staining and p21WAF1/Cip1 expression. Moreover, knockdown of iASPP induced PLK1 downregulation, whereas either genes' silencing sensitized the cells in response to cisplatin treatment. More prominent apoptosis was induced by cisplatin in OCCC cells after the knockdown of either iASPP or PLK1 as evidenced by the formation of more cleaved caspase 3. Heightened chemosensitivity to cisplatin after iASPP knockdown was further demonstrated in in vivo xenograft model. Additionally, both iASPP and PLK1 were shown to regulate autophagic flux as the induction of LC3B-II and LC3 puncta were much less in OCCC cells with either knockdown. Importantly, inhibition of autophagy also enhanced chemosensitivity to cisplatin in OCCC cells. These findings strongly imply that iASPP and PLK1 affect the chemoresistance of OCCC via the regulation of autophagy and apoptosis. Both iASPP and PLK1 can be potential therapeutic targets for treating OCCC in combination with conventional chemotherapy.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis
- Autophagy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Proliferation
- Cisplatin/pharmacology
- Drug Resistance, Neoplasm
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Prognosis
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Survival Rate
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Ka-Kui Chan
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Oscar Gee-Wan Wong
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Esther Shuk-Ying Wong
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Karen Kar-Loen Chan
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Philip Pun-Ching Ip
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Ka-Yu Tse
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
| | - Annie Nga-Yin Cheung
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong Island, Hong Kong
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Zhang XY, Zhang M, Cong Q, Zhang MX, Zhang MY, Lu YY, Xu CJ. Hexokinase 2 confers resistance to cisplatin in ovarian cancer cells by enhancing cisplatin-induced autophagy. Int J Biochem Cell Biol 2017; 95:9-16. [PMID: 29247711 DOI: 10.1016/j.biocel.2017.12.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 01/06/2023]
Abstract
The high mortality rate of ovarian cancer is connected with the development of acquired resistance to multiple cancer drugs, especially cisplatin. Activation of cytoprotective autophagy has been implicated as a contributing mechanism for acquired cisplatin resistance in ovarian cancer cells. Hexokinase 2 (HK2) phosphorylates glucose to generate glucose-6-phosphate, the rate-limiting step in glycolysis. Higher HK2 expression has been associated with chemoresistance in ovarian cancer. However, whether HK2 functionally contributes to cisplatin resistance in ovarian cancer is unclear. In this study, we investigated the role of HK2 in regulating ovarian cancer cisplatin resistance. Increased HK2 levels were detected in drug-resistant human ovarian cancer cells and tissues. Cisplatin downregulated HK2 in cisplatin-sensitive but not in resistant ovarian cancer cells. HK2 knockdown sensitized resistant ovarian cancer cells to cisplatin-induced cell death and apoptosis. Conversely, HK2 overexpression in cisplatin-sensitive cells induced cisplatin resistance. Mechanistically, cisplatin increased ERK1/2 phosphorylation as well as autophagic activity. Blocking autophagy with the autophagy inhibitor 3-MA sensitized resistant ovarian cancer cells to cisplatin. HK2 overexpression enhanced cisplatin-induced ERK1/2 phosphorylation and autophagy while HK2 knockdown showed the opposite effects. Blocking the MEK/ERK pathway using the MEK inhibitor U0126 prevented cisplatin-induced autophagy enhanced by HK2 overexpression. Furthermore, HK2 knockdown sensitized resistance ovarian tumor xenografts to cisplatin in vivo. In conclusion, our data supported that HK2 promotes cisplatin resistance in ovarian cancer by enhancing drug-induced, ERK-mediated autophagy. Therefore, targeting HK2 may be a new therapeutic strategy to combat chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Xiao-Yan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Meng Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Qing Cong
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Ming-Xing Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Meng-Yu Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Ying-Ying Lu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Cong-Jian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China.
| |
Collapse
|
20
|
Bortnik S, Gorski SM. Clinical Applications of Autophagy Proteins in Cancer: From Potential Targets to Biomarkers. Int J Mol Sci 2017; 18:ijms18071496. [PMID: 28696368 PMCID: PMC5535986 DOI: 10.3390/ijms18071496] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 12/20/2022] Open
Abstract
Autophagy, a lysosome-mediated intracellular degradation and recycling pathway, plays multiple context-dependent roles in tumorigenesis and treatment resistance. Encouraging results from various preclinical studies have led to the initiation of numerous clinical trials with the intention of targeting autophagy in various cancers. Accumulating knowledge of the particular mechanisms and players involved in different steps of autophagy regulation led to the ongoing discovery of small molecule inhibitors designed to disrupt this highly orchestrated process. However, the development of validated autophagy-related biomarkers, essential for rational selection of patients entering clinical trials involving autophagy inhibitors, is lagging behind. One possible source of biomarkers for this purpose is the autophagy machinery itself. In this review, we address the recent trends, challenges and advances in the assessment of the biomarker potential of clinically relevant autophagy proteins in human cancers.
Collapse
Affiliation(s)
- Svetlana Bortnik
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada.
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada.
| | - Sharon M Gorski
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada.
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada.
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| |
Collapse
|
21
|
DeVorkin L, Hattersley M, Kim P, Ries J, Spowart J, Anglesio MS, Levi SM, Huntsman DG, Amaravadi RK, Winkler JD, Tinker AV, Lum JJ. Autophagy Inhibition Enhances Sunitinib Efficacy in Clear Cell Ovarian Carcinoma. Mol Cancer Res 2017; 15:250-258. [PMID: 28184014 PMCID: PMC5451253 DOI: 10.1158/1541-7786.mcr-16-0132] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/24/2016] [Accepted: 11/16/2016] [Indexed: 01/07/2023]
Abstract
Clear cell ovarian carcinoma (CCOC) is an aggressive form of epithelial ovarian cancer that exhibits low response rates to systemic therapy and poor patient outcomes. Multiple studies in CCOC have revealed expression profiles consistent with increased hypoxia, and our previous data suggest that hypoxia is correlated with increased autophagy in CCOC. Hypoxia-induced autophagy is a key factor promoting tumor cell survival and resistance to therapy. Recent clinical trials with the molecular-targeted receptor tyrosine kinase (RTK) inhibitor sunitinib have demonstrated limited activity. Here, it was evaluated whether the hypoxia-autophagy axis could be modulated to overcome resistance to sunitinib. Importantly, a significant increase in autophagic activity was found with a concomitant loss in cell viability in CCOC cells treated with sunitinib. Pharmacologic inhibition of autophagy with the lysosomotropic analog Lys05 inhibited autophagy and enhanced sunitinib-mediated suppression of cell viability. These results were confirmed by siRNA targeting the autophagy-related gene Atg5 In CCOC tumor xenografts, Lys05 potentiated the antitumor activity of sunitinib compared with either treatment alone. These data reveal that CCOC tumors have an autophagic dependency and are an ideal tumor histotype for autophagy inhibition as a strategy to overcome resistance to RTK inhibitors like sunitinib.Implications: This study shows that autophagy inhibition enhances sunitinib-mediated cell death in a preclinical model of CCOC. Mol Cancer Res; 15(3); 250-8. ©2017 AACR.
Collapse
Affiliation(s)
- Lindsay DeVorkin
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Matthew Hattersley
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada
| | - Paul Kim
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada
| | - Jenna Ries
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada
| | - Jaeline Spowart
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Michael S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Samuel M Levi
- Department of Chemistry, School of Arts and Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ravi K Amaravadi
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey D Winkler
- Department of Chemistry, School of Arts and Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anna V Tinker
- BC Cancer Agency, Division of Medical Oncology, Vancouver Centre, Vancouver, British Columbia, Canada
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer Agency, Victoria, British Columbia, Canada.
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
22
|
Miyamoto M, Takano M, Aoyama T, Soyama H, Yoshikawa T, Tsuda H, Furuya K. Inhibition of autophagy protein LC3A as a therapeutic target in ovarian clear cell carcinomas. J Gynecol Oncol 2017; 28:e33. [PMID: 28382796 PMCID: PMC5391392 DOI: 10.3802/jgo.2017.28.e33] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/17/2017] [Accepted: 01/31/2017] [Indexed: 11/30/2022] Open
Abstract
Objective Ovarian clear cell carcinoma (CCC) is one of histological subtypes showing poor prognosis due to chemoresistance. The association of autophagy-related proteins and clinical implementation in CCC has not been determined. Methods The present study investigated whether expression of autophagy-related protein, light chain 3A (LC3A), was related with prognoses in the patients with CCC using immuno-histochemical stainings, and whether inhibition of autophagy modified the sensitivity to cisplatin in CCC cells in vitro. Results High expression of autophagy-related protein, LC3A, was detected in 78 cases (78%) in all CCC cases. The patients with high LC3A expression showed significantly lower response rate to primary chemotherapy (17% vs. 100%, p<0.010), and had worse progression-free survival (PFS) and overall survival (OS) compared with those with LC3A low expression. Furthermore, multivariate analyses revealed that high expression of LC3A was identified as independent worse prognostic factors for PFS and OS. Inhibition of autophagy protein LC3A using hydroxychloroquine (HCQ) increased sensitivity to cisplatin in CCC cells in vitro. Conclusion High expression of LC3A proteins was associated with lower response to platinum therapy, leading to worse prognoses in CCC. Although further studies are needed to confirm the results, inhibition of autophagy by HCQ was associated with platinum sensitivity. Autophagy protein LC3A could be a promising target for treatment for CCC.
Collapse
Affiliation(s)
- Morikazu Miyamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Masashi Takano
- Department of Clinical Oncology, National Defense Medical College Hospital, Tokorozawa, Japan.
| | - Tadashi Aoyama
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hiroaki Soyama
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Tomoyuki Yoshikawa
- Department of Clinical Oncology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Kenichi Furuya
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| |
Collapse
|
23
|
Wang YF, Xu YL, Tang ZH, Li T, Zhang LL, Chen X, Lu JH, Leung CH, Ma DL, Qiang WA, Wang YT, Lu JJ. Baicalein Induces Beclin 1- and Extracellular Signal-Regulated Kinase-Dependent Autophagy in Ovarian Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:123-136. [DOI: 10.1142/s0192415x17500094] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Baicalein (BA), one of the major compounds isolated from the root of Scutellaria baicalensis Gerogi, exhibits various pharmacological effects, such as anti-oxidant, anti-inflammatory, and anticancer effects. In this study, we found that BA reduced cell viability and increased apoptosis in ovarian cancer cells. Treatment of cells with BA enhanced microtubule-associated protein light chain 3-II (LC3-II) expression, acidic vesicular organelle and GFP-LC3 fluorescence dot accumulation. Combined treatment with chloroquine and BA apparently reduced cell viability and increased the cleavage of poly (ADPribose) polymerase (PARP) in both HEY and A2780 ovarian cancer cell lines, indicating that BA induces a protective autophagy in these cells. Knockdown of Beclin 1 by siRNA remarkably decreased BA-induced LC3-II lipidation. In addition, we found an increase in the phosphorylation of extracellular signal-regulated kinase (ERK, Thr202/Thr204) and AKT (Ser473) after BA treatment, and inhibition of ERK activation by the pharmacological inhibitor U0126 or ERK siRNA blocked BA-induced autophagy. Taken together, these results suggest that BA induces Beclin 1- and ERK-dependent autophagy in ovarian cancer cells.
Collapse
Affiliation(s)
- Ya-Fang Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Yu-Lian Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Zheng-Hai Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Le-Le Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, P.R. China
| | - Wen-An Qiang
- Department of Obstetrics and Gynecology-Division of Reproductive Science in Medicine, Department of Pathology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P.R. China
| |
Collapse
|
24
|
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: 0.3] [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
|
25
|
Koizume S, Ito S, Yoshioka Y, Kanayama T, Nakamura Y, Yoshihara M, Yamada R, Ochiya T, Ruf W, Miyagi E, Hirahara F, Miyagi Y. High-level secretion of tissue factor-rich extracellular vesicles from ovarian cancer cells mediated by filamin-A and protease-activated receptors. Thromb Haemost 2015; 115:299-310. [PMID: 26446354 DOI: 10.1160/th15-03-0213] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/14/2015] [Indexed: 12/19/2022]
Abstract
Thromboembolic events occur frequently in ovarian cancer patients. Tissue factor (TF) is often overexpressed in tumours, including ovarian clear-cell carcinoma (CCC), a subtype with a generally poor prognosis. TF-coagulation factor VII (fVII) complexes on the cell surface activate downstream coagulation mechanisms. Moreover, cancer cells secrete extracellular vesicles (EVs), which act as vehicles for TF. We therefore examined the characteristics of EVs produced by ovarian cancer cells of various histological subtypes. CCC cells secreted high levels of TF within EVs, while the high-TF expressing breast cancer cell line MDA-MB-231 shed fewer TF-positive EVs. We also found that CCC tumours with hypoxic tissue areas synthesised TF and fVII in vivo, rendering the blood of xenograft mice bearing these tumours hypercoagulable compared with mice bearing MDA-MB-231 tumours. Incorporation of TF into EVs and secretion of EVs from CCC cells exposed to hypoxia were both dependent on the actin-binding protein, filamin-A (filA). Furthermore, production of these EVs was dependent on different protease-activated receptors (PARs) on the cell surface. These results show that CCC cells could produce large numbers of TF-positive EVs dependent upon filA and PARs. This phenomenon may be the mechanism underlying the increased incidence of venous thromboembolism in ovarian cancer patients.
Collapse
Affiliation(s)
- Shiro Koizume
- Shiro Koizume, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan, Tel.: +81 45 391 5761, E-mail:
| | | | | | | | | | | | | | | | | | | | | | - Yohei Miyagi
- Yohei Miyagi, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan, Tel.: +81 45 391 5761, E-mail:
| |
Collapse
|
26
|
Abstract
Autophagy is a highly regulated catabolic process involving lysosomal degradation of intracellular components, damaged organelles, misfolded proteins, and toxic aggregates, reducing oxidative stress and protecting cells from damage. The process is also induced in response to various conditions, including nutrient deprivation, metabolic stress, hypoxia, anticancer therapeutics, and radiation therapy to adapt cellular conditions for survival. Autophagy can function as a tumor suppressor mechanism in normal cells and dysregulation of this process (ie, monoallelic Beclin-1 deletion) may lead to malignant transformation and carcinogenesis. In tumors, autophagy is thought to promote tumor growth and progression by helping cells to adapt and survive in metabolically-challenged and harsh tumor microenvironments (ie, hypoxia and acidity). Recent in vitro and in vivo studies in preclinical models suggested that modulation of autophagy can be used as a therapeutic modality to enhance the efficacy of conventional therapies, including chemo and radiation therapy. Currently, more than 30 clinical trials are investigating the effects of autophagy inhibition in combination with cytotoxic chemotherapies and targeted agents in various cancers. In this review, we will discuss the role, molecular mechanism, and regulation of autophagy, while targeting this process as a novel therapeutic modality, in various cancers.
Collapse
Affiliation(s)
- Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas - Houston, MD Anderson Cancer Center, Houston, TX, USA
| | - Doris M Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma HSC, Oklahoma City, OK, USA
| |
Collapse
|
27
|
Davidson B, Tropé CG. Ovarian cancer: diagnostic, biological and prognostic aspects. ACTA ACUST UNITED AC 2015; 10:519-33. [PMID: 25335543 DOI: 10.2217/whe.14.37] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ovarian cancer remains the most lethal gynecologic malignancy, owing to late detection, intrinsic and acquired chemoresistance and remarkable heterogeneity. Despite optimization of surgical and chemotherapy protocols and initiation of clinical trials incorporating targeted therapy, only modest gains have been achieved in prolonging survival in this cancer. This review provides an update of recent developments in our understanding of the etiology, origin, diagnosis, progression and treatment of this malignancy, with emphasis on clinically relevant genetic classification approaches. In the authors' opinion, focused effort directed at understanding the molecular make-up of recurrent and metastatic ovarian cancer, while keeping in mind the unique molecular character of each of its histological types, is central to our effort to improve patient outcome in this cancer.
Collapse
Affiliation(s)
- Ben Davidson
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310 Oslo, Norway
| | | |
Collapse
|
28
|
Kobayashi H, Sugimoto H, Onishi S, Nakano K. Novel biomarker candidates for the diagnosis of ovarian clear cell carcinoma. Oncol Lett 2015; 10:612-618. [PMID: 26622542 DOI: 10.3892/ol.2015.3367] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 05/27/2015] [Indexed: 02/07/2023] Open
Abstract
Ovarian clear cell carcinoma can arise from endometriosis; however, it is distinct from other types of epithelial ovarian carcinoma in terms of its clinicopathological and molecular features. Cancer antigen 125 lacks the sensitivity and specificity required for accurate clinical diagnosis of clear cell carcinoma. Therefore, the aim of the current review was to identify novel biomarker candidates for the immunohistochemical and serological diagnosis of clear cell carcinoma. A search of the relevant English language literature published between 1966 and 2014 was conducted using the PubMed MEDLINE online database. High-throughput tissue microarray technology and proteomic screening combined with mass spectrometry may provide additional information regarding diagnostic biomarker candidates for ovarian clear cell carcinoma. The present review summarizes the characteristics of potential genomic alterations that activate cancer signaling pathways and, thus, contribute to carcinogenesis. The major signaling pathways activated in clear cell carcinoma are associated with cell cycle regulation (hepatitis A virus cellular receptor 1 and tumor protein D52), growth factor signaling (insulin-like growth factor binding protein 1; KiSS-1 metastasis-suppressor; erb-b2 receptor tyrosine kinase 2; and fibroblast growth factor receptor 2), anti-apoptosis and survival pathways [sialidase 3 (membrane sialidase)], metabolism (γ-glutamyltransferase 1), chemoresistance (napsin A aspartic peptidase, glutathione peroxidase 3; and aldehyde dehydrogenase 1 family, member A1), coagulation [coagulation factor III (thromboplastin, tissue factor); and tissue factor pathway inhibitor 2], signaling (lectin, galactoside-binding and soluble, 3), and adhesion and the extracellular matrix [cadherin 1, type 1, E-cadherin (epithelial); versican; and laminin, α 5]. The present review of the relevant literature may provide a basis for additional clinical investigation of the ovarian clear cell carcinoma serum biomarker candidate proteins identified herein.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hitomi Sugimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Shunsuke Onishi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kazutoshi Nakano
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
29
|
Gibson SB. Autophagy in clear cell ovarian cancer, a potential marker for hypoxia and poor prognosis?(#). J Pathol 2015; 228:434-6. [PMID: 22951989 DOI: 10.1002/path.4100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 08/24/2012] [Accepted: 08/28/2012] [Indexed: 12/27/2022]
Abstract
Autophagy contributes to cell survival and is up-regulated under hypoxia in many different cancers. Ovarian cancer has a poor prognosis and is generally resistant to chemotherapy. Through genetic profiling, it has becoming evident that ovarian cancer has distinct subtypes but the significance of these subtypes in ovarian cancer remains unclear. In this issue, Dr Lum and colleagues have presented evidence that autophagy as measured by LC3A staining occurs in a clear cell ovarian cancer that is correlated with hypoxic regions and poor overall survival. In addition, autophagy under hypoxia appears to be higher in clear cell ovarian cancer cells compared to other subtypes. This indicates that autophagy could be a factor in drug resistance and poor survival in clear cell ovarian cancer patients. This insight could lead to a better understanding of the role of autophagy under hypoxia in human ovarian cancer and could be a valuable biomarker for the development of better therapies for clear cell ovarian cancers.
Collapse
Affiliation(s)
- Spencer B Gibson
- Manitoba Institute of Cell Biology, 675 McDermot Avenue, Manitoba, Canada; CancerCare Manitoba, Manitoba, Canada; Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
30
|
Kao C, Chao A, Tsai CL, Chuang WC, Huang WP, Chen GC, Lin CY, Wang TH, Wang HS, Lai CH. Bortezomib enhances cancer cell death by blocking the autophagic flux through stimulating ERK phosphorylation. Cell Death Dis 2014; 5:e1510. [PMID: 25375375 PMCID: PMC4260726 DOI: 10.1038/cddis.2014.468] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/10/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022]
Abstract
The antitumor activity of an inhibitor of 26S proteasome bortezomib (Velcade) has been observed in various malignancies, including colon cancer, prostate cancer, breast cancer, and ovarian cancer. Bortezomib has been proposed to stimulate autophagy, but scientific observations did not always support this. Interactions between ERK activity and autophagy are complex and not completely clear. Autophagy proteins have recently been shown to regulate the functions of ERK, and ERK activation has been found to induce autophagy. On the other hand, sustained activation of ERK has also been shown to inhibit the maturation step of the autophagy process. In this study, we sought to identify the mechanism of autophagy regulation in cancer cells treated with bortezomib. Our results indicate that bortezomib blocked the autophagic flux without inhibiting the fusion of the autophagosome and lysosome. In ovarian cancer, as well as endometrial cancer and hepatocellular carcinoma cells, bortezomib inhibited protein degradation in lysosomes by suppressing cathepsins, which requires the participation of ERK phosphorylation, but not JNK or p38. Our findings that ERK phosphorylation reduced cathepsins further explain how ERK phosphorylation inhibits the autophagic flux. In conclusion, bortezomib may induce ERK phosphorylation to suppress cathepsin B and inhibit the catalytic process of autophagy in ovarian cancer and other solid tumors. The inhibition of cisplatin-induced autophagy by bortezomib can enhance chemotherapy efficacy in ovarian cancer. As we also found that bortezomib blocks the autophagic flux in other cancers, the synergistic cytotoxic effect of bortezomib by abolishing chemotherapy-related autophagy may help us develop strategies of combination therapies for multiple cancers.
Collapse
Affiliation(s)
- C Kao
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - A Chao
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - C-L Tsai
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - W-C Chuang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - W-P Huang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - G-C Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - C-Y Lin
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - T-H Wang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan [3] Genomic Medicine Research Core Laboratory, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [4] School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - H-S Wang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - C-H Lai
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
31
|
Okada M, Oikawa M, Miki Y, Shimizu Y, Echigo S, Takahashi T, Kumamoto H. Immunohistochemical assessment of ATG7, LC3, and p62 in ameloblastomas. J Oral Pathol Med 2014; 43:606-12. [PMID: 24762217 DOI: 10.1111/jop.12177] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND To investigate the roles of autophagy in tumorigenesis, cytodifferentiation, and prognosis of odontogenic tumors, we analyzed the immunohistochemical expression of ATG7, LC3, and p62 in odontogenic tissues. METHODS Tissue specimens of nine dental follicles and 69 ameloblastomas were immunohistochemically examined with antibodies against ATG7, LC3, and p62. RESULTS Immunohistochemical reactivity for ATG7, LC3, and p62 was detected in many odontogenic epithelial cells and several endothelial cells and fibroblasts in dental follicles and ameloblastomas. ATG7 reactivity in ameloblatomas was significantly higher than that in dental follicles. Expression of ATG7, LC3, and p62 was found markedly in neoplastic cells near the basement membrane rather than central polyhedral cells in ameloblastomas. Reactivity for these molecules was significantly higher in unicystic ameloblastomas than in solid ameloblastomas. Granular cells in granular cell ameloblastomas showed obvious reactivity for the autophagy- related molecules, and LC3 reactivity in granular cell ameloblastomas was significantly higher than in other ameloblastoma variations. Recurrent ameloblastomas showed significantly lower reactivity of LC3 and p62 than primary ameloblastomas. CONCLUSIONS Expression of ATG7, LC3, and p62 in dental follicles and ameloblastomas suggests that autophagy regulation might be affected by microenvironment alterations during tumorigenesis. The molecular machinery for autophagy is possibly involved in tissue architecture, neoplastic cell differentiation, and prognosis of the benign epithelial odontogenic tumor.
Collapse
Affiliation(s)
- Miwa Okada
- Division of Oral and Maxillofacial Surgery, Department of Oral Medicine and Surgery, Tohoku University Graduate School of Dentistry, Sendai, Japan; Division of Oral Pathology, Department of Oral Medicine and Surgery, Tohoku Universtiy Graduate School of Dentistry, Sendai, Japan; Department of Dentistry and Oral Maxillofacial Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Kim JY, Jung WH, Koo JS. Expression of autophagy-related proteins according to androgen receptor and HER-2 status in estrogen receptor-negative breast cancer. PLoS One 2014; 9:e105666. [PMID: 25140630 PMCID: PMC4139390 DOI: 10.1371/journal.pone.0105666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 07/27/2014] [Indexed: 01/07/2023] Open
Abstract
PURPOSE The purpose of this study was to investigate the expression of autophagy-related proteins in relation to androgen receptor (AR) status in estrogen receptor (ER)-negative breast cancers. METHODS We extracted 334 ER-negative breast cancer samples to construct tissue microarrays (TMAs), which were immunohistochemically stained for autophagy-related proteins (beclin-1, LC3A, LC3B, p62) and for AR and HER-2. RESULTS There were 127 AR-positive cases and 207 AR-negative cases, and 140 HER-2-positive cases and 194 HER-2 negative cases. The AR-negative group was associated with tumoral LC3A expression (P<0.001), while the AR-positive group was associated with tumoral BNIP3 expression (P<0.001). Tumoral LC3A was most highly expressed in the AR-negative and HER-2 negative group, while stromal LC3A showed the highest expression in the AR-negative and HER-2-positive group. Tumoral BNIP3 and stromal BNIP3 were highest in the AR-positive and HER-2-negative group. In the AR-positive and HER-2-negative group, stromal p62 positivity was an independent factor that was statistically significant in its association with shorter disease-free survival (DFS) (Hazard ratio: 10.21, 95% CI: 1.130-92.31, P = 0.039). Shorter DFS was associated with tumoral LC3A positivity (Hazard ratio: 10.28, 95% CI: 2.068-51.19, P = 0.004) in the AR-negative and HER-2-positive group. CONCLUSION In ER-negative breast cancers, AR status was associated with expression of different types of autophagy-related proteins. Tumoral LC3A was most highly expressed in AR-negative breast cancers, while tumor BNIP3 was highest in AR-positive breast cancers.
Collapse
Affiliation(s)
- Ji-Ye Kim
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Hee Jung
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ja Seung Koo
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
33
|
Expression and clinical significance of the autophagy proteins BECLIN 1 and LC3 in ovarian cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:462658. [PMID: 25136588 PMCID: PMC4127242 DOI: 10.1155/2014/462658] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022]
Abstract
Autophagy is dysregulated in cancer and might be involved in ovarian carcinogenesis. BECLIN-1, a protein that interacts with either BCL-2 or PI3k class III, plays a critical role in the regulation of both autophagy and cell death. Induction of autophagy is associated with the presence of vacuoles characteristically labelled with the protein LC3. We have studied the biological and clinical significance of BECLIN 1 and LC3 in ovary tumours of different histological types. The positive expression of BECLIN 1 was well correlated with the presence of LC3-positive autophagic vacuoles and was inversely correlated with the expression of BCL-2. The latter inhibits the autophagy function of BECLIN 1. We found that type I tumours, which are less aggressive than type II, were more frequently expressing high level of BECLIN 1. Of note, tumours of histologic grade III expressed low level of BECLIN 1. Consistently, high level of expression of BECLIN 1 and LC3 in tumours is well correlated with the overall survival of the patients. The present data are compatible with the hypotheses that a low level of autophagy favours cancer progression and that ovary cancer with upregulated autophagy has a less aggressive behaviour and is more responsive to chemotherapy.
Collapse
|
34
|
Clinical significance of autophagic protein LC3 levels and its correlation with XIAP expression in hepatocellular carcinoma. Med Oncol 2014; 31:108. [DOI: 10.1007/s12032-014-0108-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/30/2014] [Indexed: 12/11/2022]
|
35
|
Prognostic significance of p62/SQSTM1 subcellular localization and LC3B in oral squamous cell carcinoma. Br J Cancer 2014; 111:944-54. [PMID: 24983366 PMCID: PMC4150268 DOI: 10.1038/bjc.2014.355] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/30/2014] [Indexed: 12/19/2022] Open
Abstract
Background: Autophagy is a programmed cell survival mechanism that has a key role in both physiologic and pathologic conditions. The relationship between autophagy and cancer is complex because autophagy can act as either a tumour suppressor or as a tumour promoter. The role of autophagy in oral squamous cell carcinoma (OSCC) is controversial. Several studies have claimed that either a high or low expression of autophagy-related proteins was associated with poor prognosis of OSCCs. The aims of the study were to compare autophagy in OSCCs, verrucous hyperplasias, and normal oral mucosas, and to inspect the prognostic role of autophagy in OSCCs. Methods: We used the autophagosome marker, LC3B, and autophagy flux marker, p62/SQSTM1 (p62), by using immunohistochemistry, and examined p62 mRNA by RNA in situ hybridization, to evaluate autophagy in 195 OSCCs, 47 verrucous hyperplasias, and 37 normal oral mucosas. The prognostic roles of LC3B and p62 protein expressions in OSCCs were investigated. Results: We discovered that the normal oral mucosa exhibited limited LC3B punctae and weak cytoplasmic p62 staining, whereas the OSCCs exhibited a marked increase in LC3B punctae and cytoplasmic p62 expression. The expression pattern of LC3B and cytoplasmic p62 of the verrucous hyperplasias were between normal oral mucosas and OSCCs. The normal oral mucosas, verrucous hyperplasias, and OSCCs presented no differences in nuclear p62 expression and the p62 mRNA level. p62 mRNA expression was elevated in a minority of cases. High p62 mRNA expression was associated with high p62 protein expression in the cytoplasm. Increased LC3B punctae, high cytoplasmic p62, and low nuclear p62 expressions in OSCCs were associated with aggressive clinicopathologic features and unfavourable prognosis. In addition, low nuclear p62 expression was an independent prognostic factor for overall and disease-specific survival rates. Furthermore, we disclosed that high cytoplasmic p62 expression accompanied with either a low or high LC3B expression, which indicated autophagy impairment under basal or activated autophagic activity, was associated with aggressive behaviour in advanced OSCCs. Conclusions: We suggested that autophagy was altered during cancer initiation and progression. Autophagy impairment contributed to cancer progression in advanced OSCCs.
Collapse
|
36
|
Liao W, Sun L, Wang C, Huang H, Liu J, Liao W, Shi M. LC3A-positive "stone-like" structures predict an adverse prognosis of gastric cancer. Anat Rec (Hoboken) 2014; 297:653-62. [PMID: 24532538 PMCID: PMC4279981 DOI: 10.1002/ar.22895] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 01/08/2014] [Accepted: 01/14/2014] [Indexed: 12/16/2022]
Abstract
Microtubule-associated protein light chain 3 (LC3A) is a reliable marker of autophagy that displays three distinct patterns of immunohistochemical staining in solid tumors: diffuse cytoplasmic staining, juxtanuclear staining, and staining of "stone-like" structures. These three patterns have a different prognostic significance in many solid tumors, but little is known about their influence in gastric cancer (GC). This study was a retrospective analysis of 188 GC patients from stages I to IV. The pattern of LC3A expression was examined in tumor and nontumor tissues by immunohistochemistry. Then, the association between the pattern of LC3A expression in GC and the prognosis was investigated by Kaplan-Meier analysis and the Cox proportional hazards model. Two distinct patterns of LC3A immunostaining (diffuse cytoplasmic expression and "stone-like" structures) were observed in GC tissues. LC3A-positive "stone-like" structures were found only in the tumors, and the number of such structures was correlated with both the tumor type and tumor stage. In addition, a high number of LC3A-positive "stone-like" structures was closely associated with an increased risk of recurrence after radical resection of stages I-III cancer (P < 0.001; HR = 0.205) and was associated with a lower overall survival rate for stage IV cancer (P < 0.001; HR = 0.364). Taken together, our data demonstrate that LC3A-positive "stone-like" structures can be used as an independent biomarker for an adverse prognosis of GC, suggesting that "stone-like" structures are correlated with the malignancy of this disease.
Collapse
Affiliation(s)
- Wenjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Matias-Guiu X, Davidson B. Prognostic biomarkers in endometrial and ovarian carcinoma. Virchows Arch 2014; 464:315-31. [PMID: 24504546 DOI: 10.1007/s00428-013-1509-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 02/06/2023]
Abstract
This article reviews the main prognostic and predictive biomarkers of endometrial (EC) and ovarian carcinoma (OC). In EC, prognosis still relies on conventional pathological features such as histological type and grade, as well as myometrial or lymphovascular space invasion. Estrogen receptor, p53, Ki-67, and ploidy analysis are the most promising biomarkers among a long list of molecules that have been proposed. Also, a number of putative predictive biomarkers have been proposed in molecular targeted therapy. In OC, prognosis is predominantly dependent on disease stage at diagnosis and the extent of residual disease at primary operation. Diagnostic markers which aid in establishing histological type in OC are available. However, not a single universally accepted predictive or prognostic marker exists to date. Targeted therapy has been growingly focused at in recent years, in view of the frequent development of chemoresistance at recurrent disease. The present review emphasizes the crucial role of correct pathological classification and stringent selection criteria of the material studied as basis for any evaluation of biological markers. It further emphasizes the promise of targeted therapy in EC and OC, while simultaneously highlighting the difficulties remaining before this can become standard of care.
Collapse
Affiliation(s)
- Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain,
| | | |
Collapse
|
38
|
Townsend KN, Spowart JE, Huwait H, Eshragh S, West NR, Elrick MA, Kalloger SE, Anglesio M, Watson PH, Huntsman DG, Lum JJ. Markers of T cell infiltration and function associate with favorable outcome in vascularized high-grade serous ovarian carcinoma. PLoS One 2013; 8:e82406. [PMID: 24376535 PMCID: PMC3871161 DOI: 10.1371/journal.pone.0082406] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/23/2013] [Indexed: 01/08/2023] Open
Abstract
Background When T cells infiltrate the tumor environment they encounter a myriad of metabolic stressors including hypoxia. Overcoming the limitations imposed by an inadequate tumor vasculature that contributes to these stressors may be a crucial step to immune cells mounting an effective anti-tumor response. We sought to determine whether the functional capacity of tumor infiltrating lymphocytes (TIL) could be influenced by the tumor vasculature and correlated this with survival in patients with ovarian cancer. Methodology and Principal Findings In 196 high-grade serous ovarian tumors, we confirmed that the tumor vascularity as measured by the marker CD31 was associated with improved patient disease-specific survival. We also found that tumors positive for markers of TIL (CD8, CD4 and forkhead box P3 (FoxP3)) and T cell function (granzyme B and T-cell restricted intracellular antigen-1 (TIA-1)) correlated significantly with elevated vascularity. In vitro, hypoxic CD8 T cells showed reduced cytolytic activity, secreted less effector cytokines and upregulated autophagy. Survival analysis revealed that patients had a significant improvement in disease-specific survival when FoxP3 expressing cells were present in CD31-high tumors compared to patients with FoxP3 expressing cells in CD31-low tumors [HR: 2.314 (95% CI 1.049–5.106); p = 0.0377]. Patients with high vascular endothelial growth factor (VEGF) expressing tumors containing granzyme B positive cells had improved survival compared to patients with granzyme B positive cells in VEGF-low tumors [HR: 2.522 (95% CI 1.097–5.799); p = 0.0294]. Significance Overall, this data provides a rationale for developing strategies aimed at improving the adaptability and function of TIL to hypoxic tumor conditions.
Collapse
Affiliation(s)
- Katelin N. Townsend
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jaeline E. Spowart
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Hassan Huwait
- Anatomical Pathology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Sima Eshragh
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nathan R. West
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Mary A. Elrick
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Steve E. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter H. Watson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - David G. Huntsman
- Centre for the Translational & Applied Genomics, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Julian J. Lum
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
- * E-mail:
| |
Collapse
|
39
|
Orfanelli T, Jeong J, Doulaveris G, Holcomb K, Witkin S. Involvement of autophagy in cervical, endometrial and ovarian cancer. Int J Cancer 2013; 135:519-28. [DOI: 10.1002/ijc.28524] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/18/2013] [Indexed: 01/06/2023]
Affiliation(s)
- T. Orfanelli
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - J.M. Jeong
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - G. Doulaveris
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - K. Holcomb
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - S.S. Witkin
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| |
Collapse
|
40
|
Anglesio MS, Wiegand KC, Melnyk N, Chow C, Salamanca C, Prentice LM, Senz J, Yang W, Spillman MA, Cochrane DR, Shumansky K, Shah SP, Kalloger SE, Huntsman DG. Type-specific cell line models for type-specific ovarian cancer research. PLoS One 2013; 8:e72162. [PMID: 24023729 PMCID: PMC3762837 DOI: 10.1371/journal.pone.0072162] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/07/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND OVARIAN CARCINOMAS CONSIST OF AT LEAST FIVE DISTINCT DISEASES: high-grade serous, low-grade serous, clear cell, endometrioid, and mucinous. Biomarker and molecular characterization may represent a more biologically relevant basis for grouping and treating this family of tumors, rather than site of origin. Molecular characteristics have become the new standard for clinical pathology, however development of tailored type-specific therapies is hampered by a failure of basic research to recognize that model systems used to study these diseases must also be stratified. Unrelated model systems do offer value for study of biochemical processes but specific cellular context needs to be applied to assess relevant therapeutic strategies. METHODS We have focused on the identification of clear cell carcinoma cell line models. A panel of 32 "ovarian cancer" cell lines has been classified into histotypes using a combination of mutation profiles, IHC mutation-surrogates, and a validated immunohistochemical model. All cell lines were identity verified using STR analysis. RESULTS Many described ovarian clear cell lines have characteristic mutations (including ARID1A and PIK3CA) and an overall molecular/immuno-profile typical of primary tumors. Mutations in TP53 were present in the majority of high-grade serous cell lines. Advanced genomic analysis of bona-fide clear cell carcinoma cell lines also support copy number changes in typical biomarkers such at MET and HNF1B and a lack of any recurrent expressed re-arrangements. CONCLUSIONS As with primary ovarian tumors, mutation status of cancer genes like ARID1A and TP53 and a general immuno-profile serve well for establishing histotype of ovarian cancer cell We describe specific biomarkers and molecular features to re-classify generic "ovarian carcinoma" cell lines into type specific categories. Our data supports the use of prototype clear cell lines, such as TOV21G and JHOC-5, and questions the use of SKOV3 and A2780 as models of high-grade serous carcinoma.
Collapse
Affiliation(s)
- Michael S. Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kimberly C. Wiegand
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nataliya Melnyk
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, University of British Columbia and Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Clara Salamanca
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leah M. Prentice
- Centre for Translational and Applied Genomics (CTAG), BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Janine Senz
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Winnie Yang
- Centre for Translational and Applied Genomics (CTAG), BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Monique A. Spillman
- Department of Obstetrics & Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Dawn R. Cochrane
- Centre for Translational and Applied Genomics (CTAG), BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Karey Shumansky
- Department of Molecular Oncology, BC Cancer Agency Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sohrab P. Shah
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Oncology, BC Cancer Agency Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Steve E. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - David G. Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Genetic Pathology Evaluation Centre, University of British Columbia and Vancouver General Hospital, Vancouver, British Columbia, Canada
- Centre for Translational and Applied Genomics (CTAG), BC Cancer Agency, Vancouver, British Columbia, Canada
| |
Collapse
|
41
|
Koukourakis MI, Giatromanolaki A, Zois CE, Sivridis E. LC3 immunostaining pitfalls. Histopathology 2013; 62:962-3. [DOI: 10.1111/his.12080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Michael I Koukourakis
- Department of Radiotherapy/Oncology; University General Hospital of Alexandroupolis; Democritus University of Thrace; Alexandroupolis; Greece
| | - Alexandra Giatromanolaki
- Department of Pathology; University General Hospital of Alexandroupolis; Democritus University of Thrace; Alexandroupolis; Greece
| | - Christos E Zois
- Department of Radiotherapy/Oncology; University General Hospital of Alexandroupolis; Democritus University of Thrace; Alexandroupolis; Greece
| | - Efthimios Sivridis
- Department of Pathology; University General Hospital of Alexandroupolis; Democritus University of Thrace; Alexandroupolis; Greece
| |
Collapse
|
42
|
Autophagy proteins in prostate cancer: relation with anaerobic metabolism and Gleason score. Urol Oncol 2013; 32:39.e11-8. [PMID: 23787295 DOI: 10.1016/j.urolonc.2013.04.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 04/06/2013] [Accepted: 04/07/2013] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Up-regulation of autophagy provides an important survival mechanism to normal and malignant cells residing in a hypoxic and unfavorable nutritional environment. Yet, its role in the biology of prostate cancer remains poorly understood. METHODS In this study we investigated the expression of four major autophagy proteins, namely the microtubule-associated protein 1 light chain 3A (LC3A), LC3B, Beclin 1, and p62, together with an enzyme of anaerobic metabolism, the lactate dehydrogenase 5 (LDH5), in relation to Gleason score and extraprostatic invasion. A series of 96 prostate adenocarcinomas was examined using immunohistochemical techniques and appropriate antibodies. RESULTS The LC3A protein was expressed in the form of "stone-like" structures, and diffuse cytoplasmic staining, the LC3B reactivity was solely cytoplasmic, whereas that of p62 and LDH5 was both cytoplasmic and nuclear. A median count of 0.90 "stone-like" structures per 200 × optical field (range 0-3.6) was highly associated with a high Gleason score. Similarly, a strong cytoplasmic LC3A, LC3B, and p62 expression, when extensive (present in>50% tumor cells per section), was significantly associated with LDH5 and a high Gleason score. In addition, extensive cytoplasmic p62 expression was related with LC3A and B reactivity and also with extraprostatic invasion. Extensive Beclin-1 expression was significantly linked with extraprostatic invasion and also with p62 and LDH5 expression. CONCLUSIONS Immunohistochemical detection of autophagy proteins may potentially prove to be useful as prognostic markers and a tool for the stratification of patients in therapeutic trials targeting autophagy in prostate cancer.
Collapse
|
43
|
Bauckman KA, Haller E, Flores I, Nanjundan M. Iron modulates cell survival in a Ras- and MAPK-dependent manner in ovarian cells. Cell Death Dis 2013; 4:e592. [PMID: 23598404 PMCID: PMC3668627 DOI: 10.1038/cddis.2013.87] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/01/2013] [Accepted: 02/19/2013] [Indexed: 02/07/2023]
Abstract
Ovarian cancer is a leading cause of cancer death in women in the United States. While the majority of ovarian cancers are serous, some rarer subtypes (i.e. clear cell) are often associated with endometriosis, a benign gynecological disease. Iron is rich in the cyst fluid of endometriosis-associated ovarian cancers and induces persistent oxidative stress. The role of iron, an essential nutrient involved in multiple cellular functions, in normal ovarian cell survival and ovarian cancer remains unclear. Iron, presented as ferric ammonium citrate (FAC), dramatically inhibits cell survival in ovarian cancer cell types associated with Ras mutations, while it is without effect in immortalized normal ovarian surface epithelial (T80) and endometriotic epithelial cells (lacking Ras mutations). Interestingly, FAC induced changes in cytoplasmic vacuolation concurrently with increases in LC3-II levels (an autophagy marker); these changes occurred in an ATG5/ATG7-dependent, beclin-1/hVps34-independent, and Ras-independent manner. Knockdown of autophagy mediators in HEY ovarian cancer cells reversed FAC-induced LC3-II levels, but there was little effect on reversing the cell death response. Intriguingly, transmission electron microscopy of FAC-treated T80 cells demonstrated abundant lysosomes (confirmed using Lysotracker) rich in iron particles, which occurred in a Ras-independent manner. Although the mitogen-activated protein kinase (MAPK) inhibitor, U0126, reversed FAC-induced LC3-II/autophagic punctae and lysosomes in a Ras-independent manner, it was remarkable that U0126 reversed cell death in malignant ovarian cells associated with Ras mutations. Moreover, FAC increased heme oxygenase-1 expression in H-Ras-overexpressing T80 cells, which was associated with increased cell death when overexpressed in T80 cells. Disruption of intracellular iron levels, via chelation of intracellular iron (deferoxamine), was also detrimental to malignant ovarian cell survival; thus, homeostatic intracellular iron levels are essential for cell survival. Collectively, our results implicate iron in modulating cell death in a Ras- and MAPK-dependent manner in ovarian cancer cells.
Collapse
Affiliation(s)
- K A Bauckman
- Moffitt Cancer Center and Research Institute, Cancer Biology Program, Tampa, FL, USA
| | - E Haller
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - I Flores
- Departments of Microbiology, Obstetrics and Gynecology, Ponce School of Medicine and Health Sciences, Ponce, Puerto Rico
| | - M Nanjundan
- Moffitt Cancer Center and Research Institute, Cancer Biology Program, Tampa, FL, USA
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
44
|
Leone RD, Amaravadi RK. Autophagy: a targetable linchpin of cancer cell metabolism. Trends Endocrinol Metab 2013; 24:209-17. [PMID: 23474062 PMCID: PMC4459128 DOI: 10.1016/j.tem.2013.01.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/28/2013] [Accepted: 01/29/2013] [Indexed: 12/25/2022]
Abstract
Cancer cells display several features of aberrant cellular metabolism. Two consequences of this dysregulated metabolism are rapid depletion of intracellular nutrients and a buildup of aggregated proteins and damaged organelles. Autophagy provides a mechanism for recycling proteins, lipids, and organelles. In cancer cells, oncogenes and conditions of severe stress drive profound upregulation of autophagy. In this setting, autophagy ameliorates the ill effects of dysregulated cellular metabolism, allowing a steady supply of nutrients and removal of damaged organelles. Although therapeutic strategies targeting cancer cell metabolism and autophagy are already entering clinical trials, further study of the precise mechanisms of interplay between oncogenic signaling, cellular metabolism, and autophagy will provide more effective strategies in the future.
Collapse
Affiliation(s)
- Robert D Leone
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|