1
|
Panda B, Tripathy A, Patra S, Kullu B, Tabrez S, Jena M. Imperative connotation of SODs in cancer: Emerging targets and multifactorial role of action. IUBMB Life 2024; 76:592-613. [PMID: 38600696 DOI: 10.1002/iub.2821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024]
Abstract
Superoxide dismutase (SOD) is a crucial enzyme responsible for the redox homeostasis inside the cell. As a part of the antioxidant defense system, it plays a pivotal role in the dismutation of the superoxide radicals (O 2 - ) generated mainly by the oxidative phosphorylation, which would otherwise bring out the redox dysregulation, leading to higher reactive oxygen species (ROS) generation and, ultimately, cell transformation, and malignancy. Several studies have shown the involvement of ROS in a wide range of human cancers. As SOD is the key enzyme in regulating ROS, any change, such as a transcriptional change, epigenetic remodeling, functional alteration, and so forth, either activates the proto-oncogenes or aberrant signaling cascades, which results in cancer. Interestingly, in some cases, SODs act as tumor promoters instead of suppressors. Furthermore, SODs have also been known to switch their role during tumor progression. In this review, we have tried to give a comprehensive account of SODs multifactorial role in various human cancers so that SODs-based therapeutic strategies could be made to thwart cancers.
Collapse
Affiliation(s)
- Biswajit Panda
- Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ankita Tripathy
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Srimanta Patra
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| | - Bandana Kullu
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mrutyunjay Jena
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| |
Collapse
|
2
|
Hashiba S, Nakano M, Yokoseki I, Takahashi E, Kondo M, Jimbo Y, Ishiguro N, Arakawa H, Fukami T, Nakajima M. Cytochrome P450 and UDP-Glucuronosyltransferase Expressions, Activities, and Induction Abilities in 3D-Cultured Human Renal Proximal Tubule Epithelial Cells. Drug Metab Dispos 2024; 52:949-956. [PMID: 38866474 DOI: 10.1124/dmd.124.001685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/17/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
The role of the kidney as an excretory organ for exogenous and endogenous compounds is well recognized, but there is a wealth of data demonstrating that the kidney has significant metabolizing capacity for a variety of exogenous and endogenous compounds that in some cases surpass the liver. The induction of drug-metabolizing enzymes by some chemicals can cause drug-drug interactions and intraindividual variability in drug clearance. In this study, we evaluated the expression and induction of cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT) isoforms in 3D-cultured primary human renal proximal tubule epithelial cells (RPTEC) to elucidate their utility as models of renal drug metabolism. CYP2B6, CYP2E1, CYP3A4, CYP3A5, and all detected UGTs (UGT1A1, UGT1A4, UGT1A6, UGT1A9, and UGT2B7) mRNA levels in 3D-RPTEC were significantly higher than those in 2D-RPTEC and HK-2 cells and were close to the levels in the human kidney cortex. CYP1B1 and CYP2J2 mRNA levels in 3D-RPTEC were comparable to those in 2D-RPTEC, HK-2 cells, and the human kidney cortex. Midazolam 1'-hydroxylation, trifluoperazine N-glucuronidation, serotonin O-glucuronidation, propofol O-glucuronidation, and morphine 3-glucuronidation in the 3D-RPTEC were significantly higher than the 2D-RPTEC and comparable to those in the HepaRG cells, although bupropion, ebastine, and calcitriol hydroxylations were not different between the 2D- and 3D-RPTEC. Treatment with ligands of the aryl hydrocarbon receptor and farnesoid X receptor induced CYP1A1 and UGT2B4 expression, respectively, in 3D-RPTEC compared with 2D-RPTEC. We provided information on the expression, activity, and induction abilities of P450s and UGTs in 3D-RPTEC as an in vitro human renal metabolism model. SIGNIFICANCE STATEMENT: This study demonstrated that the expression of cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) in 3D-cultured primary human renal proximal tubule epithelial cells (3D-RPTEC) was higher than those in 2D-cultured primary human renal proximal tubule epithelial cells and HK-2 cells. The results were comparable to that in the human kidney cortex. 3D-RPTEC are useful for evaluating the induction of kidney P450s, UDP-glucuronosyltransferases, and human renal drug metabolism in cellulo.
Collapse
Affiliation(s)
- Shiori Hashiba
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Itsuki Yokoseki
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Etsushi Takahashi
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Masayuki Kondo
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Yoichi Jimbo
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Naoki Ishiguro
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Hiroshi Arakawa
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology (S.H., Ma.N., I.Y., T.F., Mi.N.) and Pharmaceutical and Health Sciences (H.A.), Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kanazawa, Japan (Ma.N., T.F., Mi.N.); R&D Department, Precision Engineering Center, Industrial Division, Nikkiso Co., Ltd., Kanazawa, Japan (E.T., M.K., Y.J.); and Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (N.I.)
| |
Collapse
|
3
|
Wang D, Chen M, Tao Z, Du J, Tian K, Chen Z, Yu B, Chen Y, Lv L. Overexpression of Extracellular Superoxide Dismutase 3 Inhibits Cancer Cell Growth and Migration in Colorectal Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:465-474. [PMID: 39128081 PMCID: PMC11232067 DOI: 10.5152/tjg.2024.23232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
BACKGROUND/AIMS Incidence of colorectal cancer is rapidly increasing worldwide. Extracellular superoxide dismutase (EcSOD; SOD3) is an antioxidant enzyme. However, SOD3 roles in colorectal cancer progression remain uncertain. MATERIALS AND METHODS Superoxide dismutase 3 expression was evaluated, and we analyzed clinical relevance of SOD3 expression in colorectal cancer. Subsequently, SOD3 roles in colorectal cancer progression were detected by gain of function experiments. Changes in subcutaneous tumor and liver nodule size after SOD3 overexpression were examined in nude mice. The expression of proliferation marker Ki67 was assessed by immunohistochemical staining. RESULTS Supperoxide dismutase 3 was downregulated in colorectal cancer (P <.01). Downregulation of SOD3 was correlated with unfavorable outcomes (P < .05). Superoxide dismutase 3 upregulation limited the proliferative (P <.05), migrative (P <.01) and invasive actions of colorectal cancer cells (P <.01) by suppressing epithelial-mesenchymal transition. Moreover, SOD3 overexpression reduced Ki67 expression (P <.01) and blocked tumor growth (P <01) and liver metastasis (P <.001) in mouse tumor model. CONCLUSION Superoxide dismutase 3 upregulation attenuates tumor growth and liver metastasis in colorectal cancer, suggesting that SOD3 has potential diagnostic and prognostic values regarding colorectal cancer treatment.
Collapse
Affiliation(s)
- Donghua Wang
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Manyu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Zhenggui Tao
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jinghu Du
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Kui Tian
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Yu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Long Lv
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
4
|
Vilchis-Landeros MM, Vázquez-Meza H, Vázquez-Carrada M, Uribe-Ramírez D, Matuz-Mares D. Antioxidant Enzymes and Their Potential Use in Breast Cancer Treatment. Int J Mol Sci 2024; 25:5675. [PMID: 38891864 PMCID: PMC11171593 DOI: 10.3390/ijms25115675] [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: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
According to the World Health Organization (WHO), breast cancer (BC) is the deadliest and the most common type of cancer worldwide in women. Several factors associated with BC exert their effects by modulating the state of stress. They can induce genetic mutations or alterations in cell growth, encouraging neoplastic development and the production of reactive oxygen species (ROS). ROS are able to activate many signal transduction pathways, producing an inflammatory environment that leads to the suppression of programmed cell death and the promotion of tumor proliferation, angiogenesis, and metastasis; these effects promote the development and progression of malignant neoplasms. However, cells have both non-enzymatic and enzymatic antioxidant systems that protect them by neutralizing the harmful effects of ROS. In this sense, antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), thioredoxin reductase (TrxR), and peroxiredoxin (Prx) protect the body from diseases caused by oxidative damage. In this review, we will discuss mechanisms through which some enzymatic antioxidants inhibit or promote carcinogenesis, as well as the new therapeutic proposals developed to complement traditional treatments.
Collapse
Affiliation(s)
- María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Melissa Vázquez-Carrada
- Institute of Microbiology, Cluster of Excellence on Plant Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Daniel Uribe-Ramírez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, Mexico City C.P. 07738, Mexico;
| | - Deyamira Matuz-Mares
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| |
Collapse
|
5
|
Bel’skaya LV, Dyachenko EI. Oxidative Stress in Breast Cancer: A Biochemical Map of Reactive Oxygen Species Production. Curr Issues Mol Biol 2024; 46:4646-4687. [PMID: 38785550 PMCID: PMC11120394 DOI: 10.3390/cimb46050282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
This review systematizes information about the metabolic features of breast cancer directly related to oxidative stress. It has been shown those redox changes occur at all levels and affect many regulatory systems in the human body. The features of the biochemical processes occurring in breast cancer are described, ranging from nonspecific, at first glance, and strictly biochemical to hormone-induced reactions, genetic and epigenetic regulation, which allows for a broader and deeper understanding of the principles of oncogenesis, as well as maintaining the viability of cancer cells in the mammary gland. Specific pathways of the activation of oxidative stress have been studied as a response to the overproduction of stress hormones and estrogens, and specific ways to reduce its negative impact have been described. The diversity of participants that trigger redox reactions from different sides is considered more fully: glycolytic activity in breast cancer, and the nature of consumption of amino acids and metals. The role of metals in oxidative stress is discussed in detail. They can act as both co-factors and direct participants in oxidative stress, since they are either a trigger mechanism for lipid peroxidation or capable of activating signaling pathways that affect tumorigenesis. Special attention has been paid to the genetic and epigenetic regulation of breast tumors. A complex cascade of mechanisms of epigenetic regulation is explained, which made it possible to reconsider the existing opinion about the triggers and pathways for launching the oncological process, the survival of cancer cells and their ability to localize.
Collapse
Affiliation(s)
- Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| | | |
Collapse
|
6
|
Ko B, An J, Lee J, Kim K, Kim T, Park S, Chae H, Youn H. Anticancer effect of superoxide dismutase on canine mammary gland tumour in vitro. Vet Med Sci 2024; 10:e1323. [PMID: 37997503 PMCID: PMC10766058 DOI: 10.1002/vms3.1323] [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: 09/25/2022] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) have been shown to promote tumour growth and metastasis in human cell lines. The superoxide anion (•O2 - ) is produced during ROS formation and is involved in tumour cell signalling. OBJECTIVES Superoxide dismutase (SOD) has been applied to canine mammary gland tumours to investigate its antitumour effects in vitro. METHODS Cell proliferation, cell cycle cell migration assays, reverse transcription-quantitative polymerase chain reaction, and western blot analysis were performed to determine the effects of SOD on canine mammary tumour cell line. RESULTS SOD treatment resulted in anti-proliferative effects and mediated cell cycle arrest in the canine mammary gland tumour cell lines (CIPp and CIPm). It also downregulated the expression of N-cadherin and Vimentin. CONCLUSIONS The results confirmed that SOD inhibits tumour cell proliferation and migration, thus supporting the potential applications of SOD as a chemotherapeutic agent for canine mammary gland tumours.
Collapse
Affiliation(s)
- Byung‐Gee Ko
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Ju‐Hyun An
- Department of Veterinary Emergency and Critical Care Medicine and Institute of Veterinary ScienceCollege of Veterinary Medicine, Kangwon National UniversityChuncheon‐siRepublic of Korea
| | - Jeong‐Hwa Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Kyeong‐Bo Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Tae‐Hyeon Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Su‐Min Park
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Hyung‐Kyu Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| | - Hwa‐Young Youn
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
7
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [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: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
8
|
Agrawal K, Asthana S, Kumar D. Role of Oxidative Stress in Metabolic Reprogramming of Brain Cancer. Cancers (Basel) 2023; 15:4920. [PMID: 37894287 PMCID: PMC10605619 DOI: 10.3390/cancers15204920] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Brain cancer is known as one of the deadliest cancers globally. One of the causative factors is the imbalance between oxidative and antioxidant activities in the body, which is referred to as oxidative stress (OS). As part of regular metabolism, oxygen is reduced by electrons, resulting in the creation of numerous reactive oxygen species (ROS). Inflammation is intricately associated with the generation of OS, leading to the increased production and accumulation of reactive oxygen and nitrogen species (RONS). Glioma stands out as one of the most common malignant tumors affecting the central nervous system (CNS), characterized by changes in the redox balance. Brain cancer cells exhibit inherent resistance to most conventional treatments, primarily due to the distinctive tumor microenvironment. Oxidative stress (OS) plays a crucial role in the development of various brain-related malignancies, such as glioblastoma multiforme (GBM) and medulloblastoma, where OS significantly disrupts the normal homeostasis of the brain. In this review, we provide in-depth descriptions of prospective targets and therapeutics, along with an assessment of OS and its impact on brain cancer metabolism. We also discuss targeted therapies.
Collapse
Affiliation(s)
- Kirti Agrawal
- School of Health Sciences and Technology (SoHST), UPES, Dehradun 248007, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), Faridabad 121001, India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun 248007, India
| |
Collapse
|
9
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
10
|
Bengtsson Y, Demircan K, Vallon-Christersson J, Malmberg M, Saal LH, Rydén L, Borg Å, Schomburg L, Sandsveden M, Manjer J. Serum copper, zinc and copper/zinc ratio in relation to survival after breast cancer diagnosis: A prospective multicenter cohort study. Redox Biol 2023; 63:102728. [PMID: 37210781 DOI: 10.1016/j.redox.2023.102728] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND The essential trace elements copper and zinc, and their ratio (copper/zinc), are important for maintaining redox homeostasis. Previous studies suggest that these elements may impact breast cancer survival. However, no epidemiological study has so far been conducted on the potential association between copper and copper/zinc levels and survival after breast cancer diagnosis. In this study, we aimed to examine the relationship between serum copper, zinc and copper/zinc levels and survival following breast cancer diagnosis. PATIENTS AND METHODS The Sweden Cancerome Analysis Network - Breast Initiative (SCAN-B) is a population-based cohort study including multiple participating hospitals in Sweden. A total of 1998 patients diagnosed with primary invasive breast cancer were followed for approximately nine years. Serum levels of copper and zinc and their ratio at the time of diagnosis was analyzed in relation to breast cancer survival using multivariate Cox regression, yielding hazard ratios (HR) with 95% confidence intervals. RESULTS A higher copper/zinc ratio was associated with lower overall survival after breast cancer diagnosis. Comparing patients with a copper/zinc ratio in quartile 4 vs 1, the crude HR was 2.29 (1.65-3.19) (Ptrend <0.01) and the fully adjusted HR was 1.58 (1.11-2.25) (Ptrend = 0.01). No overall associations were seen between serum copper or zinc levels on their own and survival after breast cancer diagnosis, although a tendency toward lower breast cancer survival was seen for higher copper levels and lower zinc levels. CONCLUSION There is evidence that the serum copper/zinc ratio provides an independent predictive value for overall survival following breast cancer diagnosis.
Collapse
Affiliation(s)
- Ylva Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden.
| | - Kamil Demircan
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health (BIH), Biomedical Innovation Academy (BIA), Berlin, Germany
| | - Johan Vallon-Christersson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE, 22381, Lund, Sweden
| | - Martin Malmberg
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE, 22381, Lund, Sweden
| | - Lisa Rydén
- Department of Surgery, Skåne University Hospital, Malmö, Sweden; Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE, 22381, Lund, Sweden
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Berlin, Germany
| | - Malte Sandsveden
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Jonas Manjer
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
11
|
Hu Q, Chen J, Yang W, Xu M, Zhou J, Tan J, Huang T. GPX3 expression was down-regulated but positively correlated with poor outcome in human cancers. Front Oncol 2023; 13:990551. [PMID: 36845676 PMCID: PMC9947857 DOI: 10.3389/fonc.2023.990551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Cancer is a crucial public health problem and one of the leading causes of death worldwide. Previous studies have suggested that GPX3 may be involved in cancer metastasis and chemotherapy resistance. However, how GPX3 affects cancer patients' outcomes and the underlying mechanism remains unclear. Methods Sequencing data and clinical data from TCGA, GTEx, HPA, and CPTAC were used to explore the relationship between GPX3 expression and clinical features. Immunoinfiltration scores were used to assess the relationship between GPX3 and the tumor immune microenvironment. Functional enrichment analysis was used to predict the role of GPX3 in tumors. Gene mutation frequency, methylation level, and histone modification were used to predict the GPX3 expression regulation method. Breast, ovarian, colon, and gastric cancer cells were used to investigate the relationship between GPX3 expression and cancer cell metastasis, proliferation, and chemotherapy sensitivity. Results GPX3 is down-regulated in various tumor tissues, and GPX3 expression level can be used as a marker for cancer diagnosis. However, GPX3 expression is associated with higher stage and lymph node metastasis, as well as poorer prognosis. GPX3 is closely related to thyroid function and antioxidant function, and its expression may be regulated by epigenetic inheritance such as methylation modification or histone modification. In vitro experiments, GPX3 expression is associated with cancer cell sensitivity to oxidant and platinum-based chemotherapy and is involved in tumor metastasis in oxidative environments. Discussion We explored the relationship between GPX3 and clinical features, immune infiltration characteristics, migration and metastasis, and chemotherapy sensitivities of human cancers. We further investigated the potential genetic and epigenetic regulation of GPX3 in cancer. Our results suggested that GPX3 plays a complicated role in the tumor microenvironment, simultaneously promoting metastasis and chemotherapy resistance in human cancers.
Collapse
Affiliation(s)
| | | | | | - Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Tan
- *Correspondence: Tao Huang, ; Jie Tan,
| | - Tao Huang
- *Correspondence: Tao Huang, ; Jie Tan,
| |
Collapse
|
12
|
METAXAS GEORGIOSI, TSIAMBAS EVANGELOS, MARINOPOULOS SPYRIDON, SPYROPOULOU DESPOINA, MANAIOS LOUKAS, ADAMOPOULOU MARIA, FALIDAS EVANGELOS, PESCHOS DIMITRIOS, KALKANI HELEN, DIMITRAKAKIS CONSTANTINE. Epigenetic Mechanisms in Breast Adenocarcinoma: Novel DNA Methylation Patterns. CANCER DIAGNOSIS & PROGNOSIS 2022; 2:603-608. [PMID: 36340455 PMCID: PMC9628153 DOI: 10.21873/cdp.10149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/08/2022] [Indexed: 11/11/2022]
Abstract
Breast adenocarcinoma is a leading cause of death in females worldwide. A broad spectrum of genetic and epigenetic alterations has been already identified and reported in millions of examined cancerous substrates, evidence of a high-level genomic heterogeneity that characterizes these malignancies. Concerning epigenetic changes and imbalances that critically affect progression and prognosis in the corresponding patients, DNA methylation, histone modifications (acetylation), micro-RNAs (miRs) alterations and chromatin re-organization represent the main mechanisms. Referring to DNA methylation, promoter hyper-hypo methylation in critical tumour suppressor and oncogenes is implicated in normal epithelia transformation to their neoplastic and finally malignant cyto-phenotypes. The current review is focused on the different methylation patterns and mechanisms detected in breast adenocarcinoma and their impact on the corresponding groups of patient response to specific chemotherapeutic regimens and life span prognosis.
Collapse
Affiliation(s)
- GEORGIOS I. METAXAS
- Breast Unit, 1st Department of Obstetrics and Gynecology, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - SPYRIDON MARINOPOULOS
- Breast Unit, 1st Department of Obstetrics and Gynecology, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - DESPOINA SPYROPOULOU
- Department of Radiation Oncology, Medical School, University of Patras, Patras, Greece
| | - LOUKAS MANAIOS
- Department of Surgery, Bioclinic Medical Center, Athens, Greece
| | - MARIA ADAMOPOULOU
- Laboratory of Molecular Microbiology and Immunology, Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Athens, Greece
| | | | - DIMITRIOS PESCHOS
- Department of Physiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | - HELEN KALKANI
- Leucippus Technology Park, NCSR Demokritos, Athens, Greece
| | - CONSTANTINE DIMITRAKAKIS
- Breast Unit, 1st Department of Obstetrics and Gynecology, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Ji L, Wang J, Yang B, Zhu J, Wang Y, Jiao J, Zhu K, Zhang M, Zhai L, Gong T, Sun C, Qin J, Wang G. Urinary protein biomarker panel predicts esophageal squamous carcinoma from control cases and other tumors. Esophagus 2022; 19:604-616. [PMID: 35792948 DOI: 10.1007/s10388-022-00932-7] [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: 02/15/2022] [Accepted: 06/07/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Discovery of noninvasive urinary biomarkers for the early diagnosis of esophageal squamous carcinoma (ESCC). METHODS We conducted proteomic analyses of 499 human urine samples obtained from healthy individuals (n = 321) and ESCC (n = 83), bladder cancer (n = 17), breast cancer (n = 12), colorectal cancer (n = 16), lung cancer (n = 33) and thyroid cancer (n = 17) patients from multiple medical centers. Those samples were divided into a discovery set (n = 247) and an independent validation set (n = 157). RESULTS Among urinary proteins identified in the comprehensive quantitative proteomics analysis, we selected a panel of three urinary biomarkers (ANXA1, S100A8, TMEM256), and established a logistic regression model in the discovery set that can correctly classify the majority of ESCC cases in the validation sets with the area under the curve (AUC) values of 0.825. This urinary biomarker panel not only discriminates ESCC patients from healthy individuals but also differentiates ESCC from other common tumors. Notably, the panel distinguishes stage I ESCC patients from healthy individuals with AUC values of 0.886. On the analysis of stage-specific biomarkers, another combination panel of protein (ANXA1, S100A8, SOD3, TMEM256) demonstrated a good AUC value of 0.792 for stage I ESCC. CONCLUSIONS Urinary biomarker panel represents a promising auxiliary diagnostic tool for ESCC, including early-stage ESCC.
Collapse
Affiliation(s)
- Linlin Ji
- Department of Thoracic Surgery, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jianping Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Bo Yang
- Department of Thoracic Surgery, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jianping Zhu
- Department of Thoracic Surgery, Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Yini Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jiaqi Jiao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Kai Zhu
- Department of Thoracic Surgery, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China
| | - Min Zhang
- Department of Oncology, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China
| | - Liqiang Zhai
- Department of Oncology, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China
| | - Tongqing Gong
- Beijing Pineal Health Management Co., Ltd, Beijing, 102206, China
| | - Changqing Sun
- Joint Center for Translational Medicine, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Guangshun Wang
- Department of Thoracic Surgery, Baodi Clinical College, Tianjin Medical University, Tianjin, 301800, China.
| |
Collapse
|
14
|
Kuo CL, Ponneri Babuharisankar A, Lin YC, Lien HW, Lo YK, Chou HY, Tangeda V, Cheng LC, Cheng AN, Lee AYL. Mitochondrial oxidative stress in the tumor microenvironment and cancer immunoescape: foe or friend? J Biomed Sci 2022; 29:74. [PMID: 36154922 PMCID: PMC9511749 DOI: 10.1186/s12929-022-00859-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/19/2022] [Indexed: 12/07/2022] Open
Abstract
The major concept of "oxidative stress" is an excess elevated level of reactive oxygen species (ROS) which are generated from vigorous metabolism and consumption of oxygen. The precise harmonization of oxidative stresses between mitochondria and other organelles in the cell is absolutely vital to cell survival. Under oxidative stress, ROS produced from mitochondria and are the major mediator for tumorigenesis in different aspects, such as proliferation, migration/invasion, angiogenesis, inflammation, and immunoescape to allow cancer cells to adapt to the rigorous environment. Accordingly, the dynamic balance of oxidative stresses not only orchestrate complex cell signaling events in cancer cells but also affect other components in the tumor microenvironment (TME). Immune cells, such as M2 macrophages, dendritic cells, and T cells are the major components of the immunosuppressive TME from the ROS-induced inflammation. Based on this notion, numerous strategies to mitigate oxidative stresses in tumors have been tested for cancer prevention or therapies; however, these manipulations are devised from different sources and mechanisms without established effectiveness. Herein, we integrate current progress regarding the impact of mitochondrial ROS in the TME, not only in cancer cells but also in immune cells, and discuss the combination of emerging ROS-modulating strategies with immunotherapies to achieve antitumor effects.
Collapse
Affiliation(s)
- Cheng-Liang Kuo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Ananth Ponneri Babuharisankar
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Ying-Chen Lin
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Hui-Wen Lien
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Yu Kang Lo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Han-Yu Chou
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Vidhya Tangeda
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Li-Chun Cheng
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - An Ning Cheng
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan. .,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan. .,Department of Life Sciences, College of Health Sciences and Technology, National Central University, Zhongli, Taoyuan, 32001, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan. .,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
15
|
Mir S, Ormsbee Golden BD, Griess BJ, Vengoji R, Tom E, Kosmacek EA, Oberley-Deegan RE, Talmon GA, Band V, Teoh-Fitzgerald ML. Upregulation of Nox4 induces a pro-survival Nrf2 response in cancer-associated fibroblasts that promotes tumorigenesis and metastasis, in part via Birc5 induction. Breast Cancer Res 2022; 24:48. [PMID: 35836253 PMCID: PMC9281082 DOI: 10.1186/s13058-022-01548-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/30/2022] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND A pro-oxidant enzyme, NADPH oxidase 4 (Nox4) has been reported to be a critical downstream effector of TGFβ-induced myofibroblast transformation during fibrosis. While there are a small number of studies suggesting an oncogenic role of Nox4 derived from activated fibroblasts, direct evidence linking this pro-oxidant to the tumor-supporting CAF phenotype and the mechanisms involved are lacking, particularly in breast cancer. METHODS We targeted Nox4 in breast patient-derived CAFs via siRNA-mediated knockdown or administration of a pharmaceutical inhibitor (GKT137831). We also determine primary tumor growth and metastasis of implanted tumor cells using a stable Nox4-/- syngeneic mouse model. Autophagic flux of CAFs was assessed using a tandem fluorescent-tagged ptfl-LC3 plasmid via confocal microscopy analysis and determination of the expression level of autophagy markers (beclin-1 and LC3B). Nox4 overexpressing CAFs depend on the Nrf2 (nuclear factor-erythroid factor 2-related factor 2) pathway for survival. We then determined the dependency of Nox4-overexpressing CAFs on the Nrf2-mediated adaptive stress response pathway for survival. Furthermore, we investigated the involvement of Birc5 on CAF phenotype (viability and collagen contraction activity) as well as the expression level of CAF markers, FAP and αSMA. CONCLUSIONS We found that deletion of stroma Nox4 and pharmaceutically targeting its activity with GKT137831 significantly inhibited orthotopic tumor growth and metastasis of implanted E0771 and 4T1 murine mammary carcinoma cell lines in mice. More importantly, we found a significant upregulation of Nox4 expression in CAFs isolated from human breast tumors versus normal mammary fibroblasts (RMFs). Our in situ RNA hybridization analysis for Nox4 transcription on a human breast tumor microarray further support a role of this pro-oxidant in the stroma of breast carcinomas. In addition, we found that Nox4 promotes autophagy in CAFs. Moreover, we found that Nox4 promoted survival of CAFs via activation of Nrf2, a master regulator of oxidative stress response. We have further shown Birc5 is involved as a downstream modulator of Nrf2-mediated pro-survival phenotype. Together these studies indicate a role of redox signaling via the Nox4-Nrf2 pathway in tumorigenesis and metastasis of breast cancer cells by promoting autophagy and survival of CAFs.
Collapse
Affiliation(s)
- Shakeel Mir
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Briana D Ormsbee Golden
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Brandon J Griess
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Eric Tom
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Melissa Lt Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
16
|
Kamiya T, Yamaguchi Y, Oka M, Hara H. Combined action of FOXO1 and superoxide dismutase 3 promotes MDA-MB-231 cell migration. Free Radic Res 2022; 56:106-114. [PMID: 35271779 DOI: 10.1080/10715762.2022.2049770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Superoxide dismutase 3 (SOD3), one of SOD isozymes, maintains extracellular redox homeostasis through the dismutation reaction of superoxide. Loss of SOD3 in tumor cells induces oxidative stress and exacerbates tumor progression; however, interestingly, overexpression of SOD3 also promotes cell proliferation through the production of hydrogen peroxide. In this study, we investigated the functional role of SOD3 in human breast cancer MDA-MB-231 cell migration and the molecular mechanisms involved in high expression of SOD3 in MDA-MB-231 cells and human monocytic THP-1 cells. The level of histone H3 trimethylation at lysine 27 (H3K27me3), a marker of gene silencing, was decreased in 12-O-tetra-decanoylphorbol-13-acetate (TPA)-treated THP-1 cells. Also, that reduction was observed within the SOD3 promoter region. We then investigated the involvement of H3K27 demethylase JMJD3 in SOD3 induction. The induction of SOD3 and the reduction of H3K27me3 were inhibited in the presence of JMJD3 inhibitor, GSK-J4. Additionally, it was first determined that the knockdown of the transcription factor forkhead box O1 (FOXO1) significantly suppressed TPA-elicited SOD3 induction. FOXO1-mediated SOD3 downregulation was also observed in MDA-MB-231 cells, and knockdown of FOXO1 and SOD3 suppressed cell migration. Our results provide a novel insight into epigenetic regulation of SOD3 expression in tumor-associated cells, and high expression of FOXO1 and SOD3 would participate in the migration of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuji Yamaguchi
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Manami Oka
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hirokazu Hara
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
17
|
Yuksel B, Deveci Ozkan A, Aydın D, Betts Z. Evaluation of the antioxidative and genotoxic effects of sodium butyrate on breast cancer cells. Saudi J Biol Sci 2022; 29:1394-1401. [PMID: 35280546 PMCID: PMC8913555 DOI: 10.1016/j.sjbs.2021.12.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Oncogenic stimulation shows a rise in reactive oxygen species (ROS), and ROS can eventually induce carcinogenesis by causing DNA damage. In this context, this study aims to evaluate some biochemical and genotoxic changes in the control of cell death caused by NaBu (Sodium butyrate). treatment in breast cancer cells. NaBu’s impact on cell proliferation was determined via WST-1 assay. The lipid peroxidation (MDA), reduced glutathione (GSH), Nitric Oxide (NO), hydrogen peroxide (H2O2), and superoxide dismutase (SOD) enzyme levels were determined biochemically. NaBu-induced genotoxic damage was estimated via single-cell gel electrophoresis (SCGE). NaBu reduced cell viability and potentially induced GSH, but decreased SOD enzyme activity and the level of MDA and NO decreased also H2O2 decreased at different times and NaBu concentrations. Higher NaBu concentrations amplified DNA damage in MCF-7 cells compared to the control group. NaBu shows anticancer and genotoxic effects, especially through antioxidant enzymes, one of the oxidative stress parameters in breast cancer. However, the anticancer and genotoxic effects of NaBu is changed in the oxidative stress parameters with time and treatment concentration of NaBu in MCF-7 cells. Furthermore, his oxidative stress-dependent effect changes need to be clarified by further evaluation with molecular and more biochemical parameters.
Collapse
|
18
|
Sarmiento-Salinas FL, Perez-Gonzalez A, Acosta-Casique A, Ix-Ballote A, Diaz A, Treviño S, Rosas-Murrieta NH, Millán-Perez-Peña L, Maycotte P. Reactive oxygen species: Role in carcinogenesis, cancer cell signaling and tumor progression. Life Sci 2021; 284:119942. [PMID: 34506835 DOI: 10.1016/j.lfs.2021.119942] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major causes of death in the world and its global burden is expected to continue increasing. In several types of cancers, reactive oxygen species (ROS) have been extensively linked to carcinogenesis and cancer progression. However, studies have reported conflicting evidence regarding the role of ROS in cancer, mostly dependent on the cancer type or the step of the tumorigenic process. We review recent studies describing diverse aspects of the interplay of ROS with cancer in the different stages of cancer progression, with a special focus on their role in carcinogenesis, their importance for cancer cell signaling and their relationship to the most prevalent cancer risk factors.
Collapse
Affiliation(s)
- Fabiola Lilí Sarmiento-Salinas
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Andrea Perez-Gonzalez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Adilene Acosta-Casique
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Adrián Ix-Ballote
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias y Tecnologías Biomédicas, Instituto Nacional de Astrofísica, Óptica y Electrónica, Puebla, Mexico
| | - Alfonso Diaz
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Samuel Treviño
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | | | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico.
| |
Collapse
|
19
|
Emerging role of ferroptosis in breast cancer: New dawn for overcoming tumor progression. Pharmacol Ther 2021; 232:107992. [PMID: 34606782 DOI: 10.1016/j.pharmthera.2021.107992] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer has become a serious threat to women's health. Cancer progression is mainly derived from resistance to apoptosis induced by procedures or therapies. Therefore, new drugs or models that can overcome apoptosis resistance should be identified. Ferroptosis is a recently identified mode of cell death characterized by excess reactive oxygen species-induced lipid peroxidation. Since ferroptosis is distinct from apoptosis, necrosis and autophagy, its induction successfully eliminates cancer cells that are resistant to other modes of cell death. Therefore, ferroptosis may become a new direction around which to design breast cancer treatment. Unfortunately, the complete appearance of ferroptosis in breast cancer has not yet been fully elucidated. Furthermore, whether ferroptosis inducers can be used in combination with traditional anti- breast cancer drugs is still unknown. Moreover, a summary of ferroptosis in breast cancer progression and therapy is currently not available. In this review, we discuss the roles of ferroptosis-associated modulators glutathione, glutathione peroxidase 4, iron, nuclear factor erythroid-2 related factor-2, superoxide dismutases, lipoxygenase and coenzyme Q in breast cancer. Furthermore, we provide evidence that traditional drugs against breast cancer induce ferroptosis, and that ferroptosis inducers eliminate breast cancer cells. Finally, we put forward prospect of using ferroptosis inducers in breast cancer therapy, and predict possible obstacles and corresponding solutions. This review will deepen our understanding of the relationship between ferroptosis and breast cancer, and provide new insights into breast cancer-related therapeutic strategies.
Collapse
|
20
|
Impact of EcSOD Perturbations in Cancer Progression. Antioxidants (Basel) 2021; 10:antiox10081219. [PMID: 34439467 PMCID: PMC8388922 DOI: 10.3390/antiox10081219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 01/29/2023] Open
Abstract
Reactive oxygen species (ROS) are a normal byproduct of cellular metabolism and are required components in cell signaling and immune responses. However, an imbalance of ROS can lead to oxidative stress in various pathological states. Increases in oxidative stress are one of the hallmarks in cancer cells, which display an altered metabolism when compared to corresponding normal cells. Extracellular superoxide dismutase (EcSOD) is an antioxidant enzyme that catalyzes the dismutation of superoxide anion (O2−) in the extracellular environment. By doing so, this enzyme provides the cell with a defense against oxidative damage by contributing to redox balance. Interestingly, EcSOD expression has been found to be decreased in a variety of cancers, and this loss of expression may contribute to the development and progression of malignancies. In addition, recent compounds can increase EcSOD activity and expression, which has the potential for altering this redox signaling and cellular proliferation. This review will explore the role that EcSOD expression plays in cancer in order to better understand its potential as a tool for the detection, predicted outcomes and potential treatment of malignancies.
Collapse
|
21
|
Xiao L, Ge X, Yang L, Chen X, Xu Q, Rui X, Fan X, Feng L, Zhang Q, Dong M, Li W. Anticancer potential of an exopolysaccharide from Lactobacillus helveticus MB2-1 on human colon cancer HT-29 cells via apoptosis induction. Food Funct 2021; 11:10170-10181. [PMID: 33164019 DOI: 10.1039/d0fo01345e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study aimed at investigating the anticancer activity of an exopolysaccharide (EPS) isolated from Lactobacillus helveticus MB2-1. The crude EPS from L. helveticus MB2-1 (LHEPS) was fractionated into three fractions, namely LHEPS-1, LHEPS-2 and LHEPS-3. LHEPS-1 exhibited the most effective anti-proliferative activity, which was associated with a stronger inhibition rate and increased lactate dehydrogenase leakage of human colon cancer HT-29 cells. Flow cytometry analysis and colorimetric assay revealed that LHEPS-1 induced cell cycle arrest by preventing G1 to S transition and increased the apoptosis rate. Furthermore, LHEPS-1 enhanced the production of intracellular reactive oxygen species (ROS) and the activity of caspases-8/9/3, increased the levels of pro-apoptotic Bax and mitochondrial cytochrome c, while decreased the anti-apoptotic Bcl-2 level, indicating that LHEPS-1 might induce the apoptosis of HT-29 cells through a ROS-dependent pathway and a mitochondria-dependent pathway. These findings suggest that LHEPS-1 may be developed as an effective food and/or drug for the prevention and therapeutics of cancer, especially human colon cancer.
Collapse
Affiliation(s)
- Luyao Xiao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Oxidative Stress in Cancer Cell Metabolism. Antioxidants (Basel) 2021; 10:antiox10050642. [PMID: 33922139 PMCID: PMC8143540 DOI: 10.3390/antiox10050642] [Citation(s) in RCA: 319] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are important in regulating normal cellular processes whereas deregulated ROS leads to the development of a diseased state in humans including cancers. Several studies have been found to be marked with increased ROS production which activates pro-tumorigenic signaling, enhances cell survival and proliferation and drives DNA damage and genetic instability. However, higher ROS levels have been found to promote anti-tumorigenic signaling by initiating oxidative stress-induced tumor cell death. Tumor cells develop a mechanism where they adjust to the high ROS by expressing elevated levels of antioxidant proteins to detoxify them while maintaining pro-tumorigenic signaling and resistance to apoptosis. Therefore, ROS manipulation can be a potential target for cancer therapies as cancer cells present an altered redox balance in comparison to their normal counterparts. In this review, we aim to provide an overview of the generation and sources of ROS within tumor cells, ROS-associated signaling pathways, their regulation by antioxidant defense systems, as well as the effect of elevated ROS production in tumor progression. It will provide an insight into how pro- and anti-tumorigenic ROS signaling pathways could be manipulated during the treatment of cancer.
Collapse
|
23
|
Reactive oxygen species (ROS): Critical roles in breast tumor microenvironment. Crit Rev Oncol Hematol 2021; 160:103285. [DOI: 10.1016/j.critrevonc.2021.103285] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/18/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
|
24
|
Aboelella NS, Brandle C, Kim T, Ding ZC, Zhou G. Oxidative Stress in the Tumor Microenvironment and Its Relevance to Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13050986. [PMID: 33673398 PMCID: PMC7956301 DOI: 10.3390/cancers13050986] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Cancer cells are consistently under oxidative stress, as reflected by elevated basal level of reactive oxygen species (ROS), due to increased metabolism driven by aberrant cell growth. This feature has been exploited to develop therapeutic strategies that control tumor growth by modulating the oxidative stress in tumor cells. This review provides an overview of recent advances in cancer therapies targeting tumor oxidative stress, and highlights the emerging evidence implicating the effectiveness of cancer immunotherapies in intensifying tumor oxidative stress. The promises and challenges of combining ROS-inducing agents with cancer immunotherapy are also discussed. Abstract It has been well-established that cancer cells are under constant oxidative stress, as reflected by elevated basal level of reactive oxygen species (ROS), due to increased metabolism driven by aberrant cell growth. Cancer cells can adapt to maintain redox homeostasis through a variety of mechanisms. The prevalent perception about ROS is that they are one of the key drivers promoting tumor initiation, progression, metastasis, and drug resistance. Based on this notion, numerous antioxidants that aim to mitigate tumor oxidative stress have been tested for cancer prevention or treatment, although the effectiveness of this strategy has yet to be established. In recent years, it has been increasingly appreciated that ROS have a complex, multifaceted role in the tumor microenvironment (TME), and that tumor redox can be targeted to amplify oxidative stress inside the tumor to cause tumor destruction. Accumulating evidence indicates that cancer immunotherapies can alter tumor redox to intensify tumor oxidative stress, resulting in ROS-dependent tumor rejection. Herein we review the recent progresses regarding the impact of ROS on cancer cells and various immune cells in the TME, and discuss the emerging ROS-modulating strategies that can be used in combination with cancer immunotherapies to achieve enhanced antitumor effects.
Collapse
Affiliation(s)
- Nada S. Aboelella
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (N.S.A.); (C.B.); (Z.-C.D.)
- The Graduate School, Augusta University, Augusta, GA 30912, USA
| | - Caitlin Brandle
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (N.S.A.); (C.B.); (Z.-C.D.)
| | - Timothy Kim
- The Center for Undergraduate Research and Scholarship, Augusta University, Augusta, GA 30912, USA;
| | - Zhi-Chun Ding
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (N.S.A.); (C.B.); (Z.-C.D.)
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Gang Zhou
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (N.S.A.); (C.B.); (Z.-C.D.)
- The Graduate School, Augusta University, Augusta, GA 30912, USA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-4472
| |
Collapse
|
25
|
Matthiesen CL, Hu L, Torslev AS, Poulsen ET, Larsen UG, Kjaer-Sorensen K, Thomsen JS, Brüel A, Enghild JJ, Oxvig C, Petersen SV. Superoxide dismutase 3 is expressed in bone tissue and required for normal bone homeostasis and mineralization. Free Radic Biol Med 2021; 164:399-409. [PMID: 33476796 DOI: 10.1016/j.freeradbiomed.2021.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Superoxide dismutase 3 (SOD3) is an extracellular protein with the capacity to convert superoxide into hydrogen peroxide, an important secondary messenger in redox regulation. To investigate the utility of zebrafish in functional studies of SOD3 and its relevance for redox regulation, we have characterized the zebrafish orthologues; Sod3a and Sod3b. Our analyses show that both recombinant Sod3a and Sod3b express SOD activity, however, only Sod3b is able to bind heparin. Furthermore, RT-PCR analyses reveal that sod3a and sod3b are expressed in zebrafish embryos and are present primarily in separate organs in adult zebrafish, suggesting distinct functions in vivo. Surprisingly, both RT-PCR and whole mount in situ hybridization showed specific expression of sod3b in skeletal tissue. To further investigate this observation, we compared femoral bone obtained from wild-type and SOD3-/- mice to determine whether a functional difference was apparent in healthy adult mice. Here we report, that bone from SOD3-/- mice is less mineralized and characterized by significant reduction of cortical and trabecular thickness in addition to reduced mechanical strength. These analyses show that SOD3 plays a hitherto unappreciated role in bone development and homeostasis.
Collapse
Affiliation(s)
| | - Lili Hu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Ebbe T Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Ulrike G Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
26
|
Ortega E, Ballester FJ, Hernández-García A, Hernández-García S, Guerrero-Rubio MA, Bautista D, Santana MD, Gandía-Herrero F, Ruiz J. Novel organo-osmium(ii) proteosynthesis inhibitors active against human ovarian cancer cells reduce gonad tumor growth inCaenorhabditis elegans. Inorg Chem Front 2021. [DOI: 10.1039/c9qi01704f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Novel Os(ii) arene complexes with a deprotonated ppy or ppy-CHO C^N ligand have been synthesized to selectively act on cancer cells as proteosynthesis inhibitorsin vitroand exert antitumor activityin vivoinC. elegansmodels.
Collapse
Affiliation(s)
- Enrique Ortega
- Departamento de Química Inorgánica
- Universidad de Murcia
- and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca)
- E-30071 Murcia
- Spain
| | - Francisco J. Ballester
- Departamento de Química Inorgánica
- Universidad de Murcia
- and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca)
- E-30071 Murcia
- Spain
| | - Alba Hernández-García
- Departamento de Química Inorgánica
- Universidad de Murcia
- and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca)
- E-30071 Murcia
- Spain
| | - Samanta Hernández-García
- Departamento de Bioquímica y Biología Molecular A
- Unidad Docente de Biología
- Facultad de Veterinaria
- Universidad de Murcia
- E-30071 Murcia
| | - M. Alejandra Guerrero-Rubio
- Departamento de Bioquímica y Biología Molecular A
- Unidad Docente de Biología
- Facultad de Veterinaria
- Universidad de Murcia
- E-30071 Murcia
| | | | - M. Dolores Santana
- Departamento de Química Inorgánica
- Universidad de Murcia
- and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca)
- E-30071 Murcia
- Spain
| | - Fernando Gandía-Herrero
- Departamento de Bioquímica y Biología Molecular A
- Unidad Docente de Biología
- Facultad de Veterinaria
- Universidad de Murcia
- E-30071 Murcia
| | - José Ruiz
- Departamento de Química Inorgánica
- Universidad de Murcia
- and Institute for Bio-Health Research of Murcia (IMIB-Arrixaca)
- E-30071 Murcia
- Spain
| |
Collapse
|
27
|
Xue N, Zhou R, Deng M, Li Y, Hu Y, Gao L, Zhang Y, Song X, Liu J, Fan R. High serum superoxide dismutase activity improves radiation-related quality of life in patients with esophageal squamous cell carcinoma. Clinics (Sao Paulo) 2021; 76:e2226. [PMID: 33909823 PMCID: PMC8050599 DOI: 10.6061/clinics/2021/e2226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/02/2020] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors in China. Intensity-modulated radiation therapy and volume-modulated arc therapy have become the main treatments for esophageal carcinoma; however, side effects caused by radiotherapy greatly impact the quality of life in these patients. This study aimed to explore the impact of serum superoxide dismutase (SOD) levels on the prognosis of patients with ESCC undergoing radiotherapy. METHODS Patients aged between 18 and 80 years with lower-middle ESCC who underwent radiotherapy were eligible for this assessment. Adverse events, responses, treatment outcomes, and overall survival (OS) were assessed. Between 2012 and 2014, 195 patients were enrolled, of which 65 were assigned to the low- and high-SOD groups based on their serum SOD values. RESULTS The baseline characteristics were similar between the two groups, except for the T staging. Adverse events in the low-SOD group were significantly higher than those in the high-SOD group (radiation esophagitis, p=0.007; radiation pneumonitis, p=0.032; leukopenia, p=0.023; thrombocytopenia, p=0.037; anemia, p=0.041). There were no significant differences in response, treatment outcomes, or OS. CONCLUSION In conclusion, high serum SOD activity improved post-radiotherapy quality of life but did not impact the prognosis of patients with ESCC. To the best of our knowledge, this study is the first to report that serum SOD activity is associated with radiation-induced toxicity and moderately increased radiotherapeutic response in patients with ESCC undergoing radiotherapy.
Collapse
Affiliation(s)
- Nannan Xue
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 453000, P.R. China
| | - Runze Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 453000, P.R. China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yitong Li
- Department of Oncology, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Yong Hu
- Department of Radiation Oncology, Hanzhong Central Hospital, Hanzhong, Shanxi 723000, P.R. China
| | - Liang Gao
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrberger Strasse, Homburg 66421, Germany
| | - Yunbo Zhang
- Department of Oncology, Zibo Bashan Wanjie hospital, Zibo, Shandong 255000, P.R. China
| | - Xiangyu Song
- Department of Radiation Oncology, Linzhou people's hospital, Linzhou, Henan 456550, P.R. China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 453000, P.R. China
- *Corresponding authors. E-mails: /
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 453000, P.R. China
- *Corresponding authors. E-mails: /
| |
Collapse
|
28
|
Lelièvre P, Sancey L, Coll JL, Deniaud A, Busser B. The Multifaceted Roles of Copper in Cancer: A Trace Metal Element with Dysregulated Metabolism, but Also a Target or a Bullet for Therapy. Cancers (Basel) 2020; 12:E3594. [PMID: 33271772 PMCID: PMC7760327 DOI: 10.3390/cancers12123594] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
In the human body, copper (Cu) is a major and essential player in a large number of cellular mechanisms and signaling pathways. The involvement of Cu in oxidation-reduction reactions requires close regulation of copper metabolism in order to avoid toxic effects. In many types of cancer, variations in copper protein levels have been demonstrated. These variations result in increased concentrations of intratumoral Cu and alterations in the systemic distribution of copper. Such alterations in Cu homeostasis may promote tumor growth or invasiveness or may even confer resistance to treatments. Once characterized, the dysregulated Cu metabolism is pinpointing several promising biomarkers for clinical use with prognostic or predictive capabilities. The altered Cu metabolism in cancer cells and the different responses of tumor cells to Cu are strongly supporting the development of treatments to disrupt, deplete, or increase Cu levels in tumors. The metallic nature of Cu as a chemical element is key for the development of anticancer agents via the synthesis of nanoparticles or copper-based complexes with antineoplastic properties for therapy. Finally, some of these new therapeutic strategies such as chelators or ionophores have shown promising results in a preclinical setting, and others are already in the clinic.
Collapse
Affiliation(s)
- Pierre Lelièvre
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Lucie Sancey
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Jean-Luc Coll
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Aurélien Deniaud
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, 38000 Grenoble, France
| | - Benoit Busser
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
- Department of Clinical Biochemistry, Grenoble Alpes University Hospital, 38043 Grenoble, France
| |
Collapse
|
29
|
Griess B, Klinkebiel D, Kueh A, Desler M, Cowan K, Fitzgerald M, Teoh-Fitzgerald M. Association ofSOD3 promoter DNA methylation with its down-regulation in breast carcinomas. Epigenetics 2020; 15:1325-1335. [PMID: 32508251 PMCID: PMC7678930 DOI: 10.1080/15592294.2020.1777666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022] Open
Abstract
Superoxide dismutase 3 (SOD3) is a secreted antioxidant enzyme that regulates reactive oxygen species in the microenvironment. It is also a potential tumour suppressor gene that is significantly downregulated in breast cancer. We have previously shown that its mRNA expression is inversely correlated with relapse free survival in breast cancer patients. This study aimed to investigate the correlation of SOD3 promoter DNA methylation with its expression in different molecular subtypes of breast carcinoma. We found that SOD3 expression was significantly reduced in breast carcinoma samples compared to normal tissues with the lowest levels observed in Luminal B subtype. Pyrosequencing analysis showed significant increase in methylation levels in the SOD3 promoter region (-108 and -19 from the TSS) in tumours vs normal tissues (53.6% vs 25.2%). The highest degree of correlation between methylation and SOD3 expression levels was observed in Luminal B subtype (Spearman's R = -0.540, P < 0.00093). In this subtype, the -78 CpG position is the most significantly methylated site. The Spearman's coefficient analysis also indicated the most significant correlation of DNA methylation at this site with SOD3 gene expression levels in tumours vs. normal tissues (R = -0.5816, P < 6.9E-12). Moreover, copy number variation analysis of TCGA database revealed that the more aggressive Triple Negative and Her2+ subtypes had higher levels of SOD3 gene deletion. The predominantly down-regulated expression pattern of SOD3 and the various genetic and epigenetic deregulations of its expression suggest that loss of this antioxidant promotes an advantageous tumour-promoting microenvironment in breast cancer.
Collapse
Affiliation(s)
- Brandon Griess
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Klinkebiel
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alice Kueh
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michelle Desler
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kenneth Cowan
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew Fitzgerald
- College of Nursing, University of Nebraska Medical Center, Omaha, NE, USA
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
30
|
Martínez-Rey D, Carmona-Rodríguez L, Fernández-Aceñero MJ, Mira E, Mañes S. Extracellular Superoxide Dismutase, the Endothelial Basement Membrane, and the WNT Pathway: New Players in Vascular Normalization and Tumor Infiltration by T-Cells. Front Immunol 2020; 11:579552. [PMID: 33250894 PMCID: PMC7673374 DOI: 10.3389/fimmu.2020.579552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are major players in the immune-mediated control of cancer and the response to immunotherapy. In primary cancers, however, TILs are commonly absent, suggesting T-cell entry into the tumor microenvironment (TME) to be selectively restricted. Blood and lymph vessels are the first barriers that circulating T-cells must cross to reach the tumor parenchyma. Certainly, the crossing of the endothelial cell (EC) basement membrane (EC-BM)-an extracellular matrix underlying EC-is a limiting step in T-cell diapedesis. This review highlights new data suggesting the antioxidant enzyme superoxide dismutase-3 (SOD3) to be a regulator of EC-BM composition in the tumor vasculature. In the EC, SOD3 induces vascular normalization and endows the EC-BM with the capacity for the extravasation of effector T-cells into the TME, which it achieves via the WNT signaling pathway. However, when activated in tumor cells, this same pathway is reported to exclude TILs. SOD3 also regulates TIL density in primary human colorectal cancers (CRC), thus affecting the relapse rate and patient survival.
Collapse
Affiliation(s)
- Diego Martínez-Rey
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| | | | - María Jesús Fernández-Aceñero
- Department of Surgical Pathology, Fundación de Investigación Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Emilia Mira
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| |
Collapse
|
31
|
Carmona-Rodríguez L, Martínez-Rey D, Fernández-Aceñero MJ, González-Martín A, Paz-Cabezas M, Rodríguez-Rodríguez N, Pérez-Villamil B, Sáez ME, Díaz-Rubio E, Mira E, Mañes S. SOD3 induces a HIF-2α-dependent program in endothelial cells that provides a selective signal for tumor infiltration by T cells. J Immunother Cancer 2020; 8:e000432. [PMID: 32591431 PMCID: PMC7319787 DOI: 10.1136/jitc-2019-000432] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs), mainly CD8+ cytotoxic T lymphocytes (CTL), are linked to immune-mediated control of human cancers and response to immunotherapy. Tumors have nonetheless developed specific mechanisms that selectively restrict T cell entry into the tumor microenvironment. The extracellular superoxide dismutase (SOD3) is an anti-oxidant enzyme usually downregulated in tumors. We hypothesize that upregulation of SOD3 in the tumor microenvironment might be a mechanism to boost T cell infiltration by normalizing the tumor-associated endothelium. RESULTS Here we show that SOD3 overexpression in endothelial cells increased in vitro transmigration of naïve and activated CD4+ and CD8+ T cells, but not of myeloid cells. Perivascular expression of SOD3 also specifically increased CD4+ and CD8+ effector T cell infiltration into tumors and improved the effectiveness of adoptively transferred tumor-specific CD8+ T cells. SOD3-induced enhanced transmigration in vitro and tumor infiltration in vivo were not associated to upregulation of T cell chemokines such as CXCL9 or CXCL10, nor to changes in the levels of endothelial adhesion receptors such as intercellular adhesion molecule-1 (ICAM-1) or vascular cell adhesion molecule-1 (VCAM-1). Instead, SOD3 enhanced T cell infiltration via HIF-2α-dependent induction of specific WNT ligands in endothelial cells; this led to WNT signaling pathway activation in the endothelium, FOXM1 stabilization, and transcriptional induction of laminin-α4 (LAMA4), an endothelial basement membrane component permissive for T cell infiltration. In patients with stage II colorectal cancer, SOD3 was associated with increased CD8+ TIL density and disease-free survival. SOD3 expression was also linked to a T cell-inflamed gene signature using the COAD cohort from The Cancer Genome Atlas program. CONCLUSION Our findings suggest that SOD3-induced upregulation of LAMA4 in endothelial cells boosts selective tumor infiltration by T lymphocytes, thus transforming immunologically "cold" into "hot" tumors. High SOD3 levels are associated with human colon cancer infiltration by CD8+ T cells, with potential consequences for the clinical outcome of these patients. Our results also uncover a cell type-specific, distinct activity of the WNT pathway for the regulation of T cell infiltration into tumors.
Collapse
Affiliation(s)
| | - Diego Martínez-Rey
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Madrid, Spain
| | | | | | - Mateo Paz-Cabezas
- Genomics and Microarray Laboratory, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | | | - Beatriz Pérez-Villamil
- Genomics and Microarray Laboratory, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | | | - Eduardo Díaz-Rubio
- Clinical Oncology, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Emilia Mira
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Madrid, Spain
| |
Collapse
|
32
|
Kang J, Sun Y, Deng Y, Liu Q, Li D, Liu Y, Guan X, Tao Z, Wang X. Autophagy-endoplasmic reticulum stress inhibition mechanism of superoxide dismutase in the formation of calcium oxalate kidney stones. Biomed Pharmacother 2020; 121:109649. [DOI: 10.1016/j.biopha.2019.109649] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 12/17/2022] Open
|
33
|
Cheng Y, Xu T, Li S, Ruan H. GPX1, a biomarker for the diagnosis and prognosis of kidney cancer, promotes the progression of kidney cancer. Aging (Albany NY) 2019; 11:12165-12176. [PMID: 31844035 PMCID: PMC6949109 DOI: 10.18632/aging.102555] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is the most common malignant tumor of the kidney, and its diagnosis and prognosis still lack reliable biomarkers. Glutathione peroxidase 1 (GPX1) has been identified to be highly expressed in a variety of human malignancies. However, few studies have studied the expression of GPX1 and its biological functions in RCC. We attempted to assess the potential of GPX1 as a promising biomarker for RCC diagnosis and prognosis. In this study, we analyzed and explored the public cancer databases (TCGA and ONCOMINE) to conclude that GPX1 is highly expressed in RCC. Meanwhile, we evaluated the expression of GPX1 at the levels of RCC cells and tissues to verify the results of the database. Moreover, high GPX1 levels were positively correlated with short overall survival time, distant metastasis, lymphatic metastasis, and tumor stage. Receiver operating characteristic curve (ROC) analysis showed that high GPX1 levels could distinguish RCC patients from normal subjects (p < 0.0001). Kaplan-Meier curve analysis revealed that high GPX1 levels predicted shorter overall survival time (p = 0.0009). Finally, the functional roles of GPX1 were examined using a GPX1 sh-RNA knockdown method in RCC cell lines. In summary, our results suggest that GPX1 may have the potential to serve as a diagnostic and prognostic biomarker for RCC patients. Moreover, targeting GPX1 may represent as a new therapeutic strategy and direction for RCC patients.
Collapse
Affiliation(s)
- Yongbiao Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tianbo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sen Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
34
|
Huang QY, Liu GF, Qian XL, Tang LB, Huang QY, Xiong LX. Long Non-Coding RNA: Dual Effects on Breast Cancer Metastasis and Clinical Applications. Cancers (Basel) 2019; 11:E1802. [PMID: 31744046 PMCID: PMC6896003 DOI: 10.3390/cancers11111802] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
As a highly heterogeneous malignancy, breast cancer (BC) has become the most significant threat to female health. Distant metastasis and therapy resistance of BC are responsible for most of the cases of mortality and recurrence. Distant metastasis relies on an array of processes, such as cell proliferation, epithelial-to-mesenchymal transition (EMT), mesenchymal-to-epithelial transition (MET), and angiogenesis. Long non-coding RNA (lncRNA) refers to a class of non-coding RNA with a length of over 200 nucleotides. Currently, a rising number of studies have managed to investigate the association between BC and lncRNA. In this study, we summarized how lncRNA has dual effects in BC metastasis by regulating invasion, migration, and distant metastasis of BC cells. We also emphasize that lncRNA has crucial regulatory effects in the stemness and angiogenesis of BC. Clinically, some lncRNAs can regulate chemotherapy sensitivity in BC patients and may function as novel biomarkers to diagnose or predict prognosis for BC patients. The exact impact on clinical relevance deserves further study. This review can be an approach to understanding the dual effects of lncRNAs in BC, thereby linking lncRNAs to quasi-personalized treatment in the future.
Collapse
Affiliation(s)
- Qi-Yuan Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Guo-Feng Liu
- First Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Xian-Ling Qian
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- First Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Li-Bo Tang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qing-Yun Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China
| |
Collapse
|
35
|
Targeting Reactive Oxygen Species in Cancer via Chinese Herbal Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9240426. [PMID: 31583051 PMCID: PMC6754955 DOI: 10.1155/2019/9240426] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Recently, reactive oxygen species (ROS), a class of highly bioactive molecules, have been extensively studied in cancers. Cancer cells typically exhibit higher levels of basal ROS than normal cells, primarily due to their increased metabolism, oncogene activation, and mitochondrial dysfunction. This moderate increase in ROS levels facilitates cancer initiation, development, and progression; however, excessive ROS concentrations can lead to various types of cell death. Therefore, therapeutic strategies that either increase intracellular ROS to toxic levels or, conversely, decrease the levels of ROS may be effective in treating cancers via ROS regulation. Chinese herbal medicine (CHM) is a major type of natural medicine and has greatly contributed to human health. CHMs have been increasingly used for adjuvant clinical treatment of tumors. Although their mechanism of action is unclear, CHMs can execute a variety of anticancer effects by regulating intracellular ROS. In this review, we summarize the dual roles of ROS in cancers, present a comprehensive analysis of and update the role of CHM—especially its active compounds and ingredients—in the prevention and treatment of cancers via ROS regulation and emphasize precautions and strategies for the use of CHM in future research and clinical trials.
Collapse
|
36
|
Sarmiento-Salinas FL, Delgado-Magallón A, Montes-Alvarado JB, Ramírez-Ramírez D, Flores-Alonso JC, Cortés-Hernández P, Reyes-Leyva J, Herrera-Camacho I, Anaya-Ruiz M, Pelayo R, Millán-Pérez-Peña L, Maycotte P. Breast Cancer Subtypes Present a Differential Production of Reactive Oxygen Species (ROS) and Susceptibility to Antioxidant Treatment. Front Oncol 2019; 9:480. [PMID: 31231612 PMCID: PMC6568240 DOI: 10.3389/fonc.2019.00480] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022] Open
Abstract
Due to their crucial role in cell metabolism and homeostasis, alterations in mitochondrial biology and function have been related to the progression of diverse diseases including cancer. One of the consequences associated to mitochondrial dysfunction is the production of reactive oxygen species (ROS). ROS are known to have a controversial role during cancer initiation and progression and although several studies have tried to manipulate intracellular ROS levels using antioxidants or pro-oxidation conditions, it is not yet clear how to target oxidation for cancer therapy. In this study, we found differences in mitochondrial morphology in breast cancer cells when compared to a non-tumorigenic cell line and differences in mitochondrial function among breast cancer subtypes when exploring gene-expression data from the TCGA tumor dataset. Interestingly, we found increased ROS levels in triple negative breast cancer (TNBC) cell lines and a dependency on ROS for survival since antioxidant treatment induced cell death in TNBC cells but not in an estrogen receptor positive (ER+) cell line. Moreover, we identified the mitochondria as the main source of ROS in TNBC cell lines. Our results indicate a potential use for ROS as a target for therapy in the TNBC subtype which currently has the worst prognosis among all breast cancers and remains as the only breast cancer subtype which lacks a targeted therapy.
Collapse
Affiliation(s)
- Fabiola Lilí Sarmiento-Salinas
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico.,Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alam Delgado-Magallón
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico.,Departamento de Bioquímica, Instituto Tecnológico de Acapulco, Acapulco de Juárez, Mexico
| | | | - Dalia Ramírez-Ramírez
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | | | - Paulina Cortés-Hernández
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Irma Herrera-Camacho
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Maricruz Anaya-Ruiz
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | | | - Paola Maycotte
- CONACYT-Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| |
Collapse
|
37
|
Use of Antimetastatic SOD3-Mimetic Albumin as a Primer in Triple Negative Breast Cancer. JOURNAL OF ONCOLOGY 2019; 2019:3253696. [PMID: 30941174 PMCID: PMC6420975 DOI: 10.1155/2019/3253696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 12/30/2018] [Indexed: 01/28/2023]
Abstract
Of the deaths attributed to cancer, 90% are due to metastasis. Treatments that prevent or cure metastasis remain elusive. Low expression of extracellular superoxide dismutase (EcSOD or SOD3) has been associated with poor outcomes and increased metastatic potential in multiple types of cancer. Here, we characterize the antimetastatic therapeutic mechanisms of a macromolecular extracellular SOD3-mimetic polynitroxyl albumin (PNA, also known as VACNO). PNA is macromolecular human serum albumin conjugated with multiple nitroxide groups and acts as an SOD-mimetic. Here we show that PNA works as a SOD3-mimetic in a highly metastatic 4T1 mouse model of triple negative breast cancer (TNBC). In vitro, PNA dose dependently inhibited 4T1 proliferation, colony formation, and reactive oxygen species (ROS) formation. In vivo, PNA enhanced reperfusion time in the hypoxic cores of 4T1 tumors as measured by ultrasound imaging. Furthermore, PNA enhanced ultrasound signal intensity within the cores of the 4T1 tumors, indicating PNA can increase blood flow and blood volume within the hypoxic cores of tumors. Lung metastasis from 4T1 flank tumor was inhibited by PNA in the presence or absence of doxorubicin, a chemotherapy agent that produces superoxide and promotes metastasis. In a separate study, PNA increased the survival of mice with 4T1 flank tumors when used in conjunction with three standard chemotherapy drugs (paclitaxel, doxorubicin, and cyclophosphamide), as compared to treatment with chemotherapy alone. In this study, PNA-increased survival was also correlated with reduction of lung metastasis. These results support the hypothesis that PNA works through the inhibition of extracellular superoxide/ROS production leading to the conversion of 4T1 cells from a metastatic tumorigenic state to a cytostatic state. These findings support future clinical trials of PNA as an antimetastatic SOD3-mimetic drug to increase overall survival in TNBC patients.
Collapse
|
38
|
Jiang H, Wang H, De Ridder M. Targeting antioxidant enzymes as a radiosensitizing strategy. Cancer Lett 2018; 438:154-164. [PMID: 30223069 DOI: 10.1016/j.canlet.2018.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/22/2018] [Accepted: 09/01/2018] [Indexed: 12/22/2022]
Abstract
Radiotherapy represents a major anti-cancer modality and effectively kills cancer cells through generation of reactive oxygen species (ROS). However, cancer cells are commonly characterized by increased activity of ROS-scavenging enzymes in adaptation to intrinsic oxidative stress, leading to radioresistance. Abrogation of this defense network by pharmacological ROS insults therefore is shown to improve radioresponse in preclinical models; some of them are then tested in clinical trials. In this review, we address (1) the importance of ROS in radioresponse, (2) the main systems regulating redox homeostasis with a special focus on their prognostic effect and predictive role in radiotherapy, and (3) the potential radiosensitizers acting through inhibition of antioxidant enzymes.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hui Wang
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
39
|
Ichihara M, Kamiya T, Hara H, Adachi T. The MEF2A and MEF2D function as scaffold proteins that interact with HDAC1 or p300 in SOD3 expression in THP-1 cells. Free Radic Res 2018; 52:799-807. [DOI: 10.1080/10715762.2018.1475730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Mari Ichihara
- Laboratory of Clinical Pharmaceutics, Department of Biomedical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Department of Biomedical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hirokazu Hara
- Laboratory of Clinical Pharmaceutics, Department of Biomedical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuo Adachi
- Laboratory of Clinical Pharmaceutics, Department of Biomedical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
40
|
Redox control in cancer development and progression. Mol Aspects Med 2018; 63:88-98. [PMID: 29501614 DOI: 10.1016/j.mam.2018.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 02/07/2023]
Abstract
Cancer is the leading cause of death worldwide after cardiovascular diseases. This has been the case for the last few decades despite there being an increase in the number of cancer treatments. One reason for the apparent lack of drug effectiveness might be, at least in part, due to unspecificity for tumors; which often leads to substantial side effects. One way to improve the treatment of cancer is to increase the specificity of the treatment in accordance with the concept of individualized medicine. This will help to prevent further progression of an existing cancer or even to reduce the tumor burden. Alternatively it would be much more attractive and efficient to prevent the development of cancer in the first place. Therefore, it is important to understand the risk factors and the mechanisms of carcinogenesis in detail. One such risk factor, often associated with tumorigenesis and tumor progression, is an increased abundance of reactive oxygen species (ROS) arising from an imbalance of ROS-producing and -eliminating components. A surplus of ROS can induce oxidative damage of macromolecules including proteins, lipids and DNA. In contrast, ROS are essential for an adequate signal transduction and are known to regulate crucial cellular processes like cellular quiescence, differentiation and even apoptosis. Therefore, regulated ROS-formation at physiological levels can inhibit tumor formation and progression. With this review we provide an overview on the current knowledge of redox control in cancer development and progression.
Collapse
|
41
|
Zhong W, Weiss HL, Jayswal RD, Hensley PJ, Downes LM, St Clair DK, Chaiswing L. Extracellular redox state shift: A novel approach to target prostate cancer invasion. Free Radic Biol Med 2018; 117:99-109. [PMID: 29421238 PMCID: PMC5845758 DOI: 10.1016/j.freeradbiomed.2018.01.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/18/2018] [Accepted: 01/20/2018] [Indexed: 01/17/2023]
Abstract
AIM Extracellular superoxide dismutase (ECSOD) and the cysteine/glutamate transporter (Cys)/(xCT) are tumor microenvironment (TME) redox state homeostasis regulators. Altered expression of ECSOD and xCT can lead to imbalance of the TME redox state and likely have a profound effect on cancer invasion. In the present study, we investigated whether ECSOD and xCT could be therapeutic targets for prostate cancer (PCa) invasion. RESULTS Immunohistochemistry of tumor microarray PCa tissues (N = 165) with high Gleason scores indicated that xCT protein expression is significantly increased while ECSOD protein expression is significantly decreased. Metastatic PCa indicated ECSOD protein expression is significantly decreased in epithelial area whereas xCT protein expression is significantly increased in stromal area. Furthermore, inhibition of extracellular O2•- by overexpression of ECSOD or alteration of the extracellular Cys/CySS ratio by knockdown of xCT protein inhibited PCa cell invasion. Simultaneous overexpression of ECSOD and knockdown xCT inhibited PCa cell invasion more than overexpression of ECSOD or knockdown of xCT alone. In the co-culturing system, simultaneous overexpression of ECSOD and knockdown of xCT in prostate stromal WPMY-1 cells inhibited PCa cell invasiveness more than overexpression of ECSOD alone. The decrease in PCa invasion correlated with increased of extracellular H2O2 levels. Notably, overexpression of catalase in TME reversed the inhibitory effect of ECSOD on cancer cell invasion. CONCLUSION Impaired ECSOD activity and an upregulated of xCT protein expression may be clinical features of an aggressive PCa, particularly metastatic cancers and/or those with a high Gleason score. Therefore, shifting the extracellular redox state toward an oxidizing status by targeted modulation of ECSOD and xCT, in both cancer and stromal cells, may provide a greater strategy for potential therapeutic interventions of aggressive PCa.
Collapse
Affiliation(s)
- Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Heidi L Weiss
- The Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, USA
| | - Rani D Jayswal
- The Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, USA
| | | | - Laura M Downes
- College of Medicine, University of Kentucky, Lexington, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA.
| |
Collapse
|
42
|
Mira E, Carmona-Rodríguez L, Pérez-Villamil B, Casas J, Fernández-Aceñero MJ, Martínez-Rey D, Martín-González P, Heras-Murillo I, Paz-Cabezas M, Tardáguila M, Oury TD, Martín-Puig S, Lacalle RA, Fabriás G, Díaz-Rubio E, Mañes S. SOD3 improves the tumor response to chemotherapy by stabilizing endothelial HIF-2α. Nat Commun 2018; 9:575. [PMID: 29422508 PMCID: PMC5805714 DOI: 10.1038/s41467-018-03079-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/18/2018] [Indexed: 02/08/2023] Open
Abstract
One drawback of chemotherapy is poor drug delivery to tumor cells, due in part to hyperpermeability of the tumor vasculature. Extracellular superoxide dismutase (SOD3) is an antioxidant enzyme usually repressed in the tumor milieu. Here we show that specific SOD3 re-expression in tumor-associated endothelial cells (ECs) increases doxorubicin (Doxo) delivery into and chemotherapeutic effect on tumors. Enhanced SOD3 activity fostered perivascular nitric oxide accumulation and reduced vessel leakage by inducing vascular endothelial cadherin (VEC) transcription. SOD3 reduced HIF prolyl hydroxylase domain protein activity, which increased hypoxia-inducible factor-2α (HIF-2α) stability and enhanced its binding to a specific VEC promoter region. EC-specific HIF-2α ablation prevented both the SOD3-mediated increase in VEC transcription and the enhanced Doxo effect. SOD3, VEC, and HIF-2α levels correlated positively in primary colorectal cancers, which suggests a similar interconnection of these proteins in human malignancy.
Collapse
Affiliation(s)
- Emilia Mira
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Lorena Carmona-Rodríguez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Beatriz Pérez-Villamil
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Josefina Casas
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - María Jesús Fernández-Aceñero
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Diego Martínez-Rey
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Paula Martín-González
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Ignacio Heras-Murillo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Mateo Paz-Cabezas
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Manuel Tardáguila
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
- Genetics Institute, University of Florida, 2033 Mowry Road, Gainesville, FL, 32610, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Silvia Martín-Puig
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares, Calle de Melchor Fernández Almagro, 3, Madrid, 28029, Spain
| | - Rosa Ana Lacalle
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Gemma Fabriás
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - Eduardo Díaz-Rubio
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain.
| |
Collapse
|
43
|
Li M, Li X, Zhuang Y, Wang Y, Burow ME, Collins-Burow B, Xue M, Song C, Shan B. Induction of HOXA9 expression in three-dimensional organotypic culture of the Claudin-low breast cancer cells. Oncotarget 2018; 7:51503-51514. [PMID: 27409175 PMCID: PMC5239492 DOI: 10.18632/oncotarget.10491] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023] Open
Abstract
The gene expression signatures of the molecular intrinsic subtypes of breast cancer are regulated by epigenetic mechanisms such as methylation of CpG islands in gene promoters. Epigenetic codes can be regulated by the tumor microenvironment. The Claudin-low subtype is associated with triple-negative invasive ductal carcinomas in patients. Herein we explored epigenetic regulation of gene expression in the Claudin-low breast cancer cells by extracellular matrix (ECM), a key component of the tumor microenvironment. We modeled attachment to ECM using laminin rich ECM three-dimensional organotypic culture (lrECM 3D). In 2D and lrECM 3D cultures we examined expression of the homeobox (HOX) genes that epigenetically regulated in development and cancer. We demonstrated induction of the selected HOX genes in lrECM 3D culture of the Claudin-low breast cancer cells MDA-MB-231 and Hs578T. In particular activation of HOXA9 expression in lrECM 3D culture required binding of bromodomain containing 4 to the HOXA9 promoter and involved CpG hypomethylation. Our findings warrant further investigation of the ECM-regulated epigenetic coding of gene expression in the Claudin-low breast cancer.
Collapse
Affiliation(s)
- Miao Li
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xi Li
- Department of Sports Medicine and Joint Surgery, The People's Hospital of Liaoning Province, Shenyang, China
| | - Yan Zhuang
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yan Wang
- Department of Biological Engineering, Zunyi Medical College Zhuhai Campus, Zhuhai, China
| | - Matthew E Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Min Xue
- Department of Physiology, Xuzhou Medical College, Xuzhou, China
| | - Chengjie Song
- Department of Physiology, Xuzhou Medical College, Xuzhou, China
| | - Bin Shan
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, Spokane, WA, USA
| |
Collapse
|
44
|
Golden BO, Griess B, Mir S, Fitzgerald M, Kuperwasser C, Domann F, Teoh-Fitzgerald M. Extracellular superoxide dismutase inhibits hepatocyte growth factor-mediated breast cancer-fibroblast interactions. Oncotarget 2017; 8:107390-107408. [PMID: 29296173 PMCID: PMC5746075 DOI: 10.18632/oncotarget.22379] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/25/2017] [Indexed: 11/25/2022] Open
Abstract
We have previously shown tumor suppressive effects of extracellular superoxide dismutase, EcSOD in breast cancer cells. In this study, an RTK signaling array revealed an inhibitory effect of EcSOD on c-Met phosphorylation and its downstream kinase c-Abl in MDA-MB231 cells. Moreover, an extracellular protein array showed that thrombospondin 1 (TSP-1), a scavenger of the c-Met ligand, hepatocyte growth factor (HGF) is significantly up-regulated in EcSOD overexpressing cells (Ec.20). We further determined the effects of EcSOD on HGF/c-Met-mediated cancer-fibroblast interactions by co-culturing normal fibroblasts (RMF) or RMF which overexpresses HGF (RMF-HGF) with MDA-MB231 cells. We observed that while RMF-HGF significantly promoted Matrigel growth of MDA-MB231, overexpression of EcSOD inhibited the HGF-stimulated growth. Similarly, a SOD mimetic, MnTE-2-PyP, inhibited HGF-induced growth and invasion of MDA-MB231. In addition, a long-term heterotypic co-culture study not only showed that Ec.20 cells are resistant to RMF-HGF-induced invasive stimulation but RMF-HGF that were co-cultured with Ec.20 cells showed an attenuated phenotype, suggesting an oxidative-mediated reciprocal interaction between the two cell types. In addition, we demonstrated that RMF-HGF showed an up-regulation of an ROS-generating enzyme, NADPH oxidase 4 (Nox4). Targeting this pro-oxidant significantly suppressed the activated phenotype of RMF-HGF in a collagen contraction assay, suggesting that RMF-HGF contributes to the oxidative tumor microenvironment. We have further shown that scavenging ROS with EcSOD significantly inhibited RMF-HGF-stimulated orthotopic tumor growth of MDA-MB231. This study suggests the loss of EcSOD in breast cancer plays a pivotal role in promoting the HGF/c-Met-mediated cancer-fibroblast interactions.
Collapse
Affiliation(s)
- Briana Ormsbee Golden
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Brandon Griess
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shakeel Mir
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew Fitzgerald
- Department of Surgery-General Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Charlotte Kuperwasser
- Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frederick Domann
- Free Radical and Radiation Biology Program, Radiation Oncology, University of Iowa, Iowa City, IA 52241, USA
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
45
|
Griess B, Tom E, Domann F, Teoh-Fitzgerald M. Extracellular superoxide dismutase and its role in cancer. Free Radic Biol Med 2017; 112:464-479. [PMID: 28842347 PMCID: PMC5685559 DOI: 10.1016/j.freeradbiomed.2017.08.013] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022]
Abstract
Reactive oxygen species (ROS) are increasingly recognized as critical determinants of cellular signaling and a strict balance of ROS levels must be maintained to ensure proper cellular function and survival. Notably, ROS is increased in cancer cells. The superoxide dismutase family plays an essential physiological role in mitigating deleterious effects of ROS. Due to the compartmentalization of ROS signaling, EcSOD, the only superoxide dismutase in the extracellular space, has unique characteristics and functions in cellular signal transduction. In comparison to the other two intracellular SODs, EcSOD is a relatively new comer in terms of its tumor suppressive role in cancer and the mechanisms involved are less well understood. Nevertheless, the degree of differential expression of this extracellular antioxidant in cancer versus normal cells/tissues is more pronounced and prevalent than the other SODs. A significant association of low EcSOD expression with reduced cancer patient survival further suggests that loss of extracellular redox regulation promotes a conducive microenvironment that favors cancer progression. The vast array of mechanisms reported in mediating deregulation of EcSOD expression, function, and cellular distribution also supports that loss of this extracellular antioxidant provides a selective advantage to cancer cells. Moreover, overexpression of EcSOD inhibits tumor growth and metastasis, indicating a role as a tumor suppressor. This review focuses on the current understanding of the mechanisms of deregulation and tumor suppressive function of EcSOD in cancer.
Collapse
Affiliation(s)
- Brandon Griess
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Eric Tom
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Frederick Domann
- Free Radical and Radiation Biology Program, Radiation Oncology, University of Iowa, Iowa, IA 52242, United States
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
46
|
Lee JR, Roh JL, Lee SM, Park Y, Cho KJ, Choi SH, Nam SY, Kim SY. Overexpression of glutathione peroxidase 1 predicts poor prognosis in oral squamous cell carcinoma. J Cancer Res Clin Oncol 2017; 143:2257-2265. [PMID: 28653098 DOI: 10.1007/s00432-017-2466-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE Intracellular antioxidant enzymes are commonly upregulated in various cancer types and are associated with treatment outcomes. Because the relationship has rarely been examined in oral squamous cell carcinoma (OSCC), we aimed to evaluate the association between the levels of glutathione peroxidase (GPX)1, GPX4, and thioredoxin reductase (TrxR)1 expression and prognosis in patients with OSCC who underwent curative surgical resection. METHODS This study included 233 patients who underwent curative surgery for previously untreated OSCC between 2000 and 2012. Tumour GPX1, GPX4, and TrxR1 expression was evaluated by immunohistochemistry and was dichotomised to low and high values according to defined expression levels. The association between GPX1, GPX4, and TrxR1 expression and clinicopathological results was analysed. Univariate and multivariate analyses using the Cox proportional hazards model were conducted to assess the significance of differences in recurrence or survival outcomes between variables. RESULTS High GPX1, GPX4, and TrxR1 expression was observed in 99 (42.5%), 133 (57.1%), and 46 (19.7%) patients, respectively. GPX1 overexpression was significantly correlated with nodal metastasis, advanced overall stage, depth of invasion of >10 mm, high grade and perineural invasion (P < 0.05). High GPX4 expression was also related to nodal metastasis, overall advanced stage and high grade (P < 0.05). Univariate and multivariate analyses showed that increased GPX1 expression was significantly associated with poor disease-free, cancer-specific and overall survival (all P < 0.05), while increased GPX4 or TrxR1 expression was not (all P > 0.1). CONCLUSIONS Tumour GPX1 expression is a useful biomarker predictive of recurrence and survival in OSCC patients.
Collapse
Affiliation(s)
- Jae Ryung Lee
- Department of Otolaryngology, Asan Medical Centre, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Centre, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Sun Mi Lee
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yangsoon Park
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung-Ja Cho
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Ho Choi
- Department of Otolaryngology, Asan Medical Centre, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soon Yuhl Nam
- Department of Otolaryngology, Asan Medical Centre, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Centre, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| |
Collapse
|
47
|
On the Origin of Superoxide Dismutase: An Evolutionary Perspective of Superoxide-Mediated Redox Signaling. Antioxidants (Basel) 2017; 6:antiox6040082. [PMID: 29084153 PMCID: PMC5745492 DOI: 10.3390/antiox6040082] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
The field of free radical biology originated with the discovery of superoxide dismutase (SOD) in 1969. Over the last 5 decades, a plethora of research has been performed in species ranging from bacteria to mammals that has elucidated the molecular reaction, subcellular location, and specific isoforms of SOD. However, while humans have only begun to study this class of enzymes over the past 50 years, it has been estimated that these enzymes have existed for billions of years, and may be some of the original enzymes found in primitive life. As life evolved over this expanse of time, these enzymes have taken on new and different functional roles potentially in contrast to how they were originally derived. Herein, examination of the evolutionary history of these enzymes provides both an explanation and further inquiries into the modern-day role of SOD in physiology and disease.
Collapse
|
48
|
Li M, Li X, Zhuang Y, Flemington EK, Lin Z, Shan B. Induction of a novel isoform of the lncRNA HOTAIR in Claudin-low breast cancer cells attached to extracellular matrix. Mol Oncol 2017; 11:1698-1710. [PMID: 28846832 PMCID: PMC5709615 DOI: 10.1002/1878-0261.12133] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/27/2017] [Accepted: 08/11/2017] [Indexed: 01/05/2023] Open
Abstract
Elevated overexpression of the lncRNA HOTAIR mediates invasion and metastasis in breast cancer. In an apparent paradox, we observed low expression of HOTAIR in the invasive Claudin‐low MDA‐MB‐231 and Hs578T cells in two‐dimensional culture (2D). However, HOTAIR expression exhibited robust induction in laminin‐rich extracellular matrix‐based three‐dimensional organotypic culture (lrECM 3D) over that in 2D culture. Induction of HOTAIR required intact ECM signaling, namely integrin α2 and SRC kinase activity. Moreover, invasive growth was suppressed by HOTAIR‐specific siRNA. Induction of HOTAIR in lrECM 3D culture resulted from the activation of a novel isoform of HOTAIR (HOTAIR‐N) whose transcription is started from the first intron of the HOXC11 gene. The HOTAIR‐N promoter exhibited increased trimethylation of histone H3 lysine 4, a histone marker of active transcription, and binding of BRD4, a reader of transcriptionally active histone markers. Inhibition of BRD4 substantially reduced the expression of HOTAIR in lrECM 3D culture. In summary, our results indicate that HOTAIR expression is activated by BRD4 binding to a novel HOTAIR‐N promoter in Claudin‐low breast cancer cells that are attached to ECM. Induction of HOTAIR is required for invasive growth of Claudin‐low breast cancer cells in lrECM 3D culture.
Collapse
Affiliation(s)
- Miao Li
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, WA, USA
| | - Xi Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, WA, USA.,Department of Sports Medicine and Joint Surgery, The People's Hospital of Liaoning Province, Shenyang, China
| | - Yan Zhuang
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zhen Lin
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bin Shan
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, WA, USA
| |
Collapse
|
49
|
Little AC, Sulovari A, Danyal K, Heppner DE, Seward DJ, van der Vliet A. Paradoxical roles of dual oxidases in cancer biology. Free Radic Biol Med 2017; 110:117-132. [PMID: 28578013 PMCID: PMC5535817 DOI: 10.1016/j.freeradbiomed.2017.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
Dysregulated oxidative metabolism is a well-recognized aspect of cancer biology, and many therapeutic strategies are based on targeting cancers by altering cellular redox pathways. The NADPH oxidases (NOXes) present an important enzymatic source of biological oxidants, and the expression and activation of several NOX isoforms are frequently dysregulated in many cancers. Cell-based studies have demonstrated a role for several NOX isozymes in controlling cell proliferation and/or cell migration, further supporting a potential contributing role for NOX in promoting cancer. While various NOX isoforms are often upregulated in cancers, paradoxical recent findings indicate that dual oxidases (DUOXes), normally prominently expressed in epithelial lineages, are frequently suppressed in epithelial-derived cancers by epigenetic mechanisms, although the functional relevance of such DUOX silencing has remained unclear. This review will briefly summarize our current understanding regarding the importance of reactive oxygen species (ROS) and NOXes in cancer biology, and focus on recent observations indicating the unique and seemingly opposing roles of DUOX enzymes in cancer biology. We will discuss current knowledge regarding the functional properties of DUOX, and recent studies highlighting mechanistic consequences of DUOX1 loss in lung cancer, and its consequences for tumor invasiveness and current anticancer therapy. Finally, we will also discuss potentially unique roles for the DUOX maturation factors. Overall, a better understanding of mechanisms that regulate DUOX and the functional consequences of DUOX silencing in cancer may offer valuable new diagnostic insights and novel therapeutic opportunities.
Collapse
Affiliation(s)
- Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States
| | - Arvis Sulovari
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States; Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States.
| |
Collapse
|
50
|
Aydin E, Johansson J, Nazir FH, Hellstrand K, Martner A. Role of NOX2-Derived Reactive Oxygen Species in NK Cell-Mediated Control of Murine Melanoma Metastasis. Cancer Immunol Res 2017; 5:804-811. [PMID: 28760732 DOI: 10.1158/2326-6066.cir-16-0382] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/04/2017] [Accepted: 07/20/2017] [Indexed: 11/16/2022]
Abstract
The NADPH oxidase of myeloid cells, NOX2, generates reactive oxygen species (ROS) to eliminate pathogens and malignant cells. NOX2-derived ROS have also been proposed to dampen functions of natural killer (NK) cells and other antineoplastic lymphocytes in the microenvironment of established tumors. The mechanisms by which NOX2 and ROS influence the process of distant metastasis have only been partially explored. Here, we utilized genetically NOX2-deficient mice and pharmacologic inhibition of NOX2 to elucidate the role of NOX2 for the hematogenous metastasis of melanoma cells. After intravenous inoculation of B16F1 or B16F10 cells, lung metastasis formation was reduced in B6.129S6-Cybbtm1DinK (Nox2-KO) versus Nox2-sufficient wild-type (WT) mice. Systemic treatment with the NOX2-inhibitor histamine dihydrochloride (HDC) reduced melanoma metastasis and enhanced the infiltration of IFNγ-producing NK cells into lungs of WT but not of Nox2-KO mice. IFNγ-deficient B6.129S7-Ifngtm1Ts /J mice were prone to develop melanoma metastases and did not respond to in vivo treatment with HDC. We propose that NOX2-derived ROS facilitate metastasis of melanoma cells by downmodulating NK-cell function and that inhibition of NOX2 may restore IFNγ-dependent, NK cell-mediated clearance of melanoma cells. Cancer Immunol Res; 5(9); 804-11. ©2017 AACR.
Collapse
Affiliation(s)
- Ebru Aydin
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Junko Johansson
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Faisal Hayat Nazir
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|