1
|
Li A, Qin Y, Gong G. The Changes of Mitochondria during Aging and Regeneration. Adv Biol (Weinh) 2024; 8:e2300445. [PMID: 38979843 DOI: 10.1002/adbi.202300445] [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: 12/04/2023] [Revised: 05/30/2024] [Indexed: 07/10/2024]
Abstract
Aging and regeneration are opposite cellular processes. Aging refers to progressive dysfunction in most cells and tissues, and regeneration refers to the replacement of damaged or dysfunctional cells or tissues with existing adult or somatic stem cells. Various studies have shown that aging is accompanied by decreased regenerative abilities, indicating a link between them. The performance of any cellular process needs to be supported by the energy that is majorly produced by mitochondria. Thus, mitochondria may be a link between aging and regeneration. It should be interesting to discuss how mitochondria behave during aging and regeneration. The changes of mitochondria in aging and regeneration discussed in this review can provide a timely and necessary study of the causal roles of mitochondrial homeostasis in longevity and health.
Collapse
Affiliation(s)
- Anqi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Guohua Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
2
|
Bi YW, Li LS, Ru N, Zhang B, Lei X. Nicotinamide adenine dinucleotide phosphate oxidase in pancreatic diseases: Mechanisms and future perspectives. World J Gastroenterol 2024; 30:429-439. [PMID: 38414585 PMCID: PMC10895600 DOI: 10.3748/wjg.v30.i5.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
Pancreatitis and pancreatic cancer (PC) stand as the most worrisome ailments affecting the pancreas. Researchers have dedicated efforts to unraveling the mechanisms underlying these diseases, yet their true nature continues to elude their grasp. Within this realm, oxidative stress is often believed to play a causal and contributory role in the development of pancreatitis and PC. Excessive accumulation of reactive oxygen species (ROS) can cause oxidative stress, and the key enzyme responsible for inducing ROS production in cells is nicotinamide adenine dinucleotide phosphate hydrogen oxides (NOX). NOX contribute to pancreatic fibrosis and inflammation by generating ROS that injure acinar cells, activate pancreatic stellate cells, and mediate macrophage polarization. Excessive ROS production occurs during malignant transformation and pancreatic carcinogenesis, creating an oxidative microenvironment that can cause abnormal apoptosis, epithelial to mesenchymal transition and genomic instability. Therefore, understanding the role of NOX in pancreatic diseases contributes to a more in-depth exploration of the exact pathogenesis of these diseases. In this review, we aim to summarize the potential roles of NOX and its mechanism in pancreatic disorders, aiming to provide novel insights into understanding the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Ya-Wei Bi
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Long-Song Li
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Nan Ru
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Zhang
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao Lei
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
4
|
Lopez-Blazquez C, Lacalle-Gonzalez C, Sanz-Criado L, Ochieng’ Otieno M, Garcia-Foncillas J, Martinez-Useros J. Iron-Dependent Cell Death: A New Treatment Approach against Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:14979. [PMID: 37834426 PMCID: PMC10573128 DOI: 10.3390/ijms241914979] [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: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating tumor type where a very high proportion of people diagnosed end up dying from cancer. Surgical resection is an option for only about 20% of patients, where the 5-year survival increase ranges from 10 to 25%. In addition to surgical resection, there are adjuvant chemotherapy schemes, such as FOLFIRINOX (a mix of Irinotecan, oxaliplatin, 5-Fluorouraci and leucovorin) or gemcitabine-based treatment. These last two drugs have been compared in the NAPOLI-3 clinical trial, and the NALIRIFOX arm was found to have a higher overall survival (OS) (11.1 months vs. 9.2 months). Despite these exciting improvements, PDAC still has no effective treatment. An interesting approach would be to drive ferroptosis in PDAC cells. A non-apoptotic reactive oxygen species (ROS)-dependent cell death, ferroptosis was first described by Dixon et al. in 2012. ROS are constantly produced in the tumor cell due to high cell metabolism, which is even higher when exposed to chemotherapy. Tumor cells have detoxifying mechanisms, such as Mn-SOD or the GSH-GPX system. However, when a threshold of ROS is exceeded in the tumor cell, the cell's antioxidant systems are overwhelmed, resulting in lipid peroxidation and, ultimately, ferroptosis. In this review, we point out ferroptosis as an approach to consider in PDAC and propose that altering the cellular ROS balance by combining oxidizing agents or with inhibitors of the main cellular detoxifiers triggers ferroptosis in PDAC.
Collapse
Affiliation(s)
- Carlos Lopez-Blazquez
- Translational Oncology Division, OncoHealth Institute, Health Research Institute—Fundación Jimenéz Diaz, Fundación Jimenéz Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), 28040 Madrid, Spain; (C.L.-B.); (L.S.-C.)
| | - Carlos Lacalle-Gonzalez
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
| | - Lara Sanz-Criado
- Translational Oncology Division, OncoHealth Institute, Health Research Institute—Fundación Jimenéz Diaz, Fundación Jimenéz Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), 28040 Madrid, Spain; (C.L.-B.); (L.S.-C.)
| | - Michael Ochieng’ Otieno
- Translational Oncology Division, OncoHealth Institute, Health Research Institute—Fundación Jimenéz Diaz, Fundación Jimenéz Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), 28040 Madrid, Spain; (C.L.-B.); (L.S.-C.)
| | - Jesus Garcia-Foncillas
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute—Fundación Jimenéz Diaz, Fundación Jimenéz Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), 28040 Madrid, Spain; (C.L.-B.); (L.S.-C.)
- Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Madrid, Spain
| |
Collapse
|
5
|
Liu WJ, Wang L, Zhou FM, Liu SW, Wang W, Zhao EJ, Yao QJ, Li W, Zhao YQ, Shi Z, Qiu JG, Jiang BH. Elevated NOX4 promotes tumorigenesis and acquired EGFR-TKIs resistance via enhancing IL-8/PD-L1 signaling in NSCLC. Drug Resist Updat 2023; 70:100987. [PMID: 37392558 DOI: 10.1016/j.drup.2023.100987] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023]
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been widely used for human non-small-cell lung cancer (NSCLC) treatment. However, acquired resistance to EGFR-TKIs is the major barrier of treatment success, and new resistance mechanism remains to be elucidated. In this study, we found that elevated NADPH oxidase 4 (NOX4) expression was associated with acquired EGFR-TKIs resistance. Gefitinib is the first-generation FDA-approved EGFR-TKI, and osimertinib is the third-generation FDA-approved EGFR-TKI. We demonstrated that NOX4 knockdown in the EGFR-TKI resistant cells enabled the cells to become sensitive to gefitinib and osimertinib treatment, while forced expression of NOX4 in the sensitive parental cells was sufficient to induce resistance to gefitinib and osimertinib in the cells. To elucidate the mechanism of NOX4 upregulation in increasing TKIs resistance, we found that knockdown of NOX4 significantly down-regulated the expression of transcription factor YY1. YY1 bound directly to the promoter region of IL-8 to transcriptionally activate IL-8 expression. Interestingly, knockdown of NOX4 and IL-8 decreased programmed death ligand 1 (PD-L1) expression, which provide new insight on TKIs resistance and immune escape. We found that patients with higher NOX4 and IL-8 expression levels showed a shorter survival time compared to those with lower NOX4 and IL-8 expression levels in response to the anti-PD-L1 therapy. Knockdown of NOX4, YY1 or IL-8 alone inhibited angiogenesis and tumor growth. Furthermore, the combination of NOX4 inhibitor GKT137831 and gefitinib had synergistic effect to inhibit cell proliferation and tumor growth and to increase cellular apoptosis. These findings demonstrated that NOX4 and YY1 were essential for mediating the acquired EGFR-TKIs resistance. IL-8 and PD-L1 are two downstream targets of NOX4 to regulate TKIs resistance and immunotherapy. These molecules may be used as potential new biomarkers and therapeutic targets for overcoming TKIs resistance in the future.
Collapse
Affiliation(s)
- Wen-Jing Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Lin Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Feng-Mei Zhou
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Shu-Wen Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Wei Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Er-Jiang Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Quan-Jun Yao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Wei Li
- Department of Pathology, Affiliated Drum Tower Hospital Nanjing University Medical School, Nanjing 210000, China
| | - Yan-Qiu Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jian-Ge Qiu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China.
| | - Bing-Hua Jiang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou 450008, China.
| |
Collapse
|
6
|
Nguyen TM, Ngoc DTM, Choi JH, Lee CH. Unveiling the Neural Environment in Cancer: Exploring the Role of Neural Circuit Players and Potential Therapeutic Strategies. Cells 2023; 12:1996. [PMID: 37566075 PMCID: PMC10417274 DOI: 10.3390/cells12151996] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
The regulation of the immune environment within the tumor microenvironment has provided new opportunities for cancer treatment. However, an important microenvironment surrounding cancer that is often overlooked despite its significance in cancer progression is the neural environment surrounding the tumor. The release of neurotrophic factors from cancer cells is implicated in cancer growth and metastasis by facilitating the infiltration of nerve cells into the tumor microenvironment. This nerve-tumor interplay can elicit cancer cell proliferation, migration, and invasion in response to neurotransmitters. Moreover, it is possible that cancer cells could establish a network resembling that of neurons, allowing them to communicate with one another through neurotransmitters. The expression levels of players in the neural circuits of cancers could serve as potential biomarkers for cancer aggressiveness. Notably, the upregulation of certain players in the neural circuit has been linked to poor prognosis in specific cancer types such as breast cancer, pancreatic cancer, basal cell carcinoma, and stomach cancer. Targeting these players with inhibitors holds great potential for reducing the morbidity and mortality of these carcinomas. However, the efficacy of anti-neurogenic agents in cancer therapy remains underexplored, and further research is necessary to evaluate their effectiveness as a novel approach for cancer treatment. This review summarizes the current knowledge on the role of players in the neural circuits of cancers and the potential of anti-neurogenic agents for cancer therapy.
Collapse
Affiliation(s)
- Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Dinh Thi Minh Ngoc
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Jung-Hye Choi
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| |
Collapse
|
7
|
Therapeutic strategies for liver diseases based on redox control systems. Biomed Pharmacother 2022; 156:113764. [DOI: 10.1016/j.biopha.2022.113764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
|
8
|
Crystallographic mining of ASK1 regulators to unravel the intricate PPI interfaces for the discovery of small molecule. Comput Struct Biotechnol J 2022; 20:3734-3754. [PMID: 35891784 PMCID: PMC9294202 DOI: 10.1016/j.csbj.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Protein seldom performs biological activities in isolation. Understanding the protein–protein interactions’ physical rewiring in response to pathological conditions or pathogen infection can help advance our comprehension of disease etiology, progression, and pathogenesis, which allow us to explore the alternate route to control the regulation of key target interactions, timely and effectively. Nonalcoholic steatohepatitis (NASH) is now a global public health problem exacerbated due to the lack of appropriate treatments. The most advanced anti-NASH lead compound (selonsertib) is withdrawn, though it is able to inhibit its target Apoptosis signal-regulating kinase 1 (ASK1) completely, indicating the necessity to explore alternate routes rather than complete inhibition. Understanding the interaction fingerprints of endogenous regulators at the molecular level that underpin disease formation and progression may spur the rationale of designing therapeutic strategies. Based on our analysis and thorough literature survey of the various key regulators and PTMs, the current review emphasizes PPI-based drug discovery’s relevance for NASH conditions. The lack of structural detail (interface sites) of ASK1 and its regulators makes it challenging to characterize the PPI interfaces. This review summarizes key regulators interaction fingerprinting of ASK1, which can be explored further to restore the homeostasis from its hyperactive states for therapeutics intervention against NASH.
Collapse
Key Words
- ASK1
- ASK1, Apoptosis signal-regulating kinase 1
- CFLAR, CASP8 and FADD-like apoptosis regulator
- CREG, Cellular repressor of E1A-stimulated genes
- DKK3, Dickkopf-related protein 3
- Interaction fingerprint
- NAFLD, Non-alcoholic fatty liver disease
- NASH
- NASH, Nonalcoholic steatohepatitis
- PPI, Protein-protein interaction
- PTM, Post-trancriptional modification
- PTMs
- Protein-protein interaction
- TNFAIP3, TNF Alpha Induced Protein 3
- TRAF2/6, Tumor necrosis factor receptor (TNFR)-associated factor2/6
- TRIM48, Tripartite Motif Containing 48
- TRX, Thioredoxin
- USP9X, Ubiquitin Specific Peptidase 9 X-Linked
Collapse
|
9
|
Gong S, Wang S, Shao M. NADPH Oxidase 4: A Potential Therapeutic Target of Malignancy. Front Cell Dev Biol 2022; 10:884412. [PMID: 35646942 PMCID: PMC9130727 DOI: 10.3389/fcell.2022.884412] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/27/2022] [Indexed: 01/05/2023] Open
Abstract
Reactive oxygen species (ROS) play a crucial role in the regulation of tumor occurrence and development. As a main source of ROS, NADPH oxidases are key enzymes that mediate electron transport within intracellular membranes. Of the NOX members that have been reported to be dysregulated in a wide variety of tumors, NOX4 is the member to be most frequently expressed. Numerous studies have elucidated that NOX4 gets involved in the regulation of tumor proliferation, metastasis, therapy resistance, tumor-stromal interaction and dysregulated tumor metabolism. In this review, we primarily discussed the biological function of NOX4 in tumorigenesis and progression of multiple cancer models, including its role in activating oncogenic signaling pathways, rewiring the metabolic phenotype and mediating immune response. Besides, the development of NOX4 inhibitors has also been unraveled. Herein, we discussed the interplay between NOX4 and tumorigenesis, proposing NOX4 as a promising therapeutic target waiting for further exploration.
Collapse
Affiliation(s)
- Shulei Gong
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shiyang Wang
- Department of Geriatric Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingrui Shao
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Mingrui Shao,
| |
Collapse
|
10
|
Potential Key Markers for Predicting the Prognosis of Gastric Adenocarcinoma Based on the Expression of Ferroptosis-Related lncRNA. J Immunol Res 2022; 2022:1249290. [PMID: 35528617 PMCID: PMC9076347 DOI: 10.1155/2022/1249290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 12/24/2022] Open
Abstract
Background. Gastric cancer is one of the most common malignant tumors, and it ranks third in global cancer-related mortality. This research was aimed at identifying new targeted treatments for gastric adenocarcinoma by constructing a ferroptosis-related lncRNA prognostic feature model. Methods. The gene expression profile and clinical data of gastric adenocarcinoma patients were downloaded from TCGA database. FerrDb database was used to determine the expression of iron death-related genes. We used R software to clean the TCAG gastric adenocarcinoma gene expression cohort and screen iron death-related differential genes and lncRNAs. The potential prognostic markers and immune infiltration characteristics were determined by constructing prognostic model and multivariate validation of lncRNA related to ferroptosis prognosis. Finally, the characteristics of immune infiltration were determined by immune correlation analysis. Results. We identified 26 ferroptosis-related lncRNAs with independent prognostic value. The Kaplan-Meier analysis identified high-risk lncRNAs associated with poor prognosis of STAD. The risk scoring model constructed by AC115619.1, AC005165.1, LINC01614, and AC002451.1 was better than traditional clinicopathological features. The 1-, 3-, and 5-year survival rates of STAD patients were predicted by the nomogram. GSEA reveals the oxidative respiration and tumor-related pathways in different risk groups. Immune analysis found significant differences in the expression of immune checkpoint-related genes TNFSF9, TNFSF4, and PDCD1LG2 between the two groups of patients. Meanwhile, there were significant differences in APC co stimulation, CCR, and checkpoint between the two groups. Conclusion. Based on the prognostic characteristics of ferroptosis-related lncRNAs, we identified the potential ferroptosis-related lncRNAs and immune infiltration characteristics in gastric adenocarcinoma, which will help provide new targeted treatments for gastric adenocarcinoma.
Collapse
|
11
|
Bekhet OH, Eid ME. The interplay between reactive oxygen species and antioxidants in cancer progression and therapy: a narrative review. Transl Cancer Res 2022; 10:4196-4206. [PMID: 35116715 PMCID: PMC8799102 DOI: 10.21037/tcr-21-629] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
Objective To unveil the role of reactive oxygen species (ROS) and antioxidants in signaling and involvement in cancer progression and therapy. Background Cancer is considered one of the main causes of mortality in developed countries and expected to be more in developing countries as well. Although some cancers may develop at young age, yet almost all types of cancers are an accumulation of genetic and epigenetic cell damages. Cancer is considered a diverse collection of diseases on a cellular level rather than a single disease; and each disease has a different cause as well. ROS have been seen as harmful toxic molecules; however, they are recognized for cellular signaling capabilities. Elevated levels of ROS have protumorigenic activities; they induce cancer cell proliferation, and adaptation to hypoxia in addition to other effects like DNA damage and genetic instability. They are produced excessively by cancer cells to hyperactivate cellular transformation meanwhile increasing antioxidant capacity to avoid cell death. Methods We discussed peer reviewed published research work from 1987 to 2021. In this paper, we review the role of antioxidants as defensive barrier against excessive ROS levels for maintaining oxidation-reduction (redox) balance; however, antioxidant can also strive in tumor cells with their scavenging capacities and maintain protumorigenic signaling and resist the cancer cell oxidative stress and apoptosis. High doses of antioxidant compounds could be toxic to cells as they are capable of reacting with the physiological concentrations of ROS present for normal cellular processes and signaling. Conclusions Maintaining cellular redox homeostasis is vital for healthy biological system. Therefore, therapeutic modalities for cancer including antioxidants and ROS management should be used at certain doses to target specific redox pathways involved in cancer progression without disrupting the overall redox balance in normal cells.
Collapse
Affiliation(s)
- Osama Hussein Bekhet
- Pole of Endocrinology, Diabetes and Nutrition, Catholic University of Louvain, Woluwe-Saint-Lambert, Belgium
| | - Mohamed Elsayed Eid
- Laboratory of Natural Products Chemistry, Mediterranean Agronomic Institute of Chania, Crete, Greece
| |
Collapse
|
12
|
Lyu PW, Xu XD, Zong K, Qiu XG. Overexpression of DUOX2 mediates doxorubicin resistance and predicts prognosis of pancreatic cancer. Gland Surg 2022; 11:115-124. [PMID: 35242674 PMCID: PMC8825507 DOI: 10.21037/gs-21-776] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/11/2022] [Indexed: 01/27/2024]
Abstract
BACKGROUND Dysregulation of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) family is frequently observed in cancers and associated with their development and progression. However, the expression, role, and clinical significance of the NOX family members in pancreatic cancer remain unexplored. METHODS The expression levels of the 7 NOX family genes were analyzed in Gene Expression Omnibus (GEO) datasets. The messenger RNA (mRNA) expression and gene alterations were explored using The Cancer Genome Atlas (TCGA) data portal. Clinical significance analyses of the NOX family genes were conducted among pancreatic cancer patients. The expression and prognostic value of dual oxidase 2 (DUOX2) were then validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC) in an independent validation cohort. The function of DUOX2 was analyzed by gene set enrichment analysis (GSEA) and its effect on the chemosensitivity of pancreatic cancer cells was detected by Cell Counting Kit-8 (CCK-8) assay. RESULTS Results showed that NOX1, NOX2 (CYBB), NOX4, DUOX1, and DUOX2 were upregulated, while NOX3 and NOX5 were downregulated in pancreatic cancer tissues compared with nontumor tissues. Genomic alteration analysis demonstrated that deregulation of NOX family genes was partially caused by genomic alterations. Survival analyses showed that only DUOX2 was associated with overall survival (OS) and relapse-free survival (RFS) of pancreatic cancer patients. The DUOX2 gene was observed to be markedly overexpressed in pancreatic cancer. In the GSEA results for pancreatic cancer patients, DUOX2 was significantly associated with oxidoreductase activity acting on nicotinamide adenine dinucleotide hydrogen (NADH) or NADPH and uridine 5'-diphospho-glucuronosyltansferase (UDP) glycosyltransferase activity. Knockdown of DUOX2 in pancreatic cancer cells increased their sensitivity to doxorubicin. CONCLUSIONS Overexpression of DUOX2 is correlated with prognosis and recurrence in pancreatic cancer patients and acts as a good marker for pancreatic cancer course prediction; furthermore, DUOX2 might be a therapeutic target for pancreatic cancer patients.
Collapse
Affiliation(s)
- Peng-Wei Lyu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Dong Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Zong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin-Guang Qiu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Bi Y, Lei X, Chai N, Linghu E. NOX4: a potential therapeutic target for pancreatic cancer and its mechanism. J Transl Med 2021; 19:515. [PMID: 34930338 PMCID: PMC8686284 DOI: 10.1186/s12967-021-03182-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) is one of the seven isoforms of NOX family, which is upregulated in pancreatic cancer cell, mouse model of pancreatic cancer and human pancreatic cancer tissue. NOX4 is a constitutively active enzyme that primarily produces hydrogen peroxide, which exhibits completely different properties from other subtypes of NOX family. More importantly, recent studies illuminate that NOX4 promotes pancreatic cancer occurrence and development in different ways. This review summarizes the potential roles and its mechanism of NOX4 in pancreatic cancer and explores NOX4 as the potential therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Yawei Bi
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Xiao Lei
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100859, China
| | - Ningli Chai
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
14
|
Mohammadi E, Tahmoorespur M, Benfeitas R, Altay O, Javadmanesh A, Lam S, Mardinoglu A, Sekhavati MH. Improvement of the performance of anticancer peptides using a drug repositioning pipeline. Biotechnol J 2021; 17:e2100417. [PMID: 34657375 DOI: 10.1002/biot.202100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 01/10/2023]
Abstract
The use of anticancer peptides (ACPs) as an alternative/complementary strategy to conventional chemotherapy treatments has been shown to decrease drug resistance and/or severe side effects. However, the efficacy of the positively-charged ACP is inhibited by elevated levels of negatively-charged cell-surface components which trap the peptides and prevent their contact with the cell membrane. Consequently, this decreases ACP-mediated membrane pore formation and cell lysis. Negatively-charged heparan sulphate (HS) and chondroitin sulphate (CS) have been shown to inhibit the cytotoxic effect of ACPs. In this study, we propose a strategy to promote the broad utilization of ACPs. In this context, we developed a drug repositioning pipeline to analyse transcriptomics data generated for four different cancer cell lines (A549, HEPG2, HT29, and MCF7) treated with hundreds of drugs in the LINCS L1000 project. Based on previous studies identifying genes modulating levels of the glycosaminoglycans (GAGs) HS and CS at the cell surface, our analysis aimed at identifying drugs inhibiting genes correlated with high HS and CS levels. As a result, we identified six chemicals as likely repositionable drugs with the potential to enhance the performance of ACPs. The codes in R and Python programming languages are publicly available in https://github.com/ElyasMo/ACPs_HS_HSPGs_CS. As a conclusion, these six drugs are highlighted as excellent targets for synergistic studies with ACPs aimed at lowering the costs associated with ACP-treatment.
Collapse
Affiliation(s)
- Elyas Mohammadi
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden.,3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ozlem Altay
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Ali Javadmanesh
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Simon Lam
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Adil Mardinoglu
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden.,Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | | |
Collapse
|
15
|
Wu S, Li T, Liu W, Huang Y. Ferroptosis and Cancer: Complex Relationship and Potential Application of Exosomes. Front Cell Dev Biol 2021; 9:733751. [PMID: 34568341 PMCID: PMC8455874 DOI: 10.3389/fcell.2021.733751] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Cell death induction has become popular as a novel cancer treatment. Ferroptosis, a newly discovered form of cell death, features regulated, iron-dependent accumulation of lipid hydroperoxides. Since this word “ferroptosis” was coined, numerous studies have examined the complex relationship between ferroptosis and cancer. Here, starting from the intrinsic hallmarks of cancer and cell death, we discuss the theoretical basis of cell death induction as a cancer treatment. We review various aspects of the relationship between ferroptosis and cancer, including the genetic basis, epigenetic modification, cancer stem cells, and the tumor microenvironment, to provide information and support for further research on ferroptosis. We also note that exosomes can be applied in ferroptosis-based therapy. These extracellular vesicles can deliver different molecules to modulate cancer cells and cell death pathways. Using exosomes to control ferroptosis occurring in targeted cells is promising for cancer therapy.
Collapse
Affiliation(s)
- Shuang Wu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tianye Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yongye Huang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
16
|
Vermot A, Petit-Härtlein I, Smith SME, Fieschi F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants (Basel) 2021; 10:890. [PMID: 34205998 PMCID: PMC8228183 DOI: 10.3390/antiox10060890] [Citation(s) in RCA: 260] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
The reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX) was first identified in the membrane of phagocytic cells. For many years, its only known role was in immune defense, where its ROS production leads to the destruction of pathogens by the immune cells. NOX from phagocytes catalyzes, via one-electron trans-membrane transfer to molecular oxygen, the production of the superoxide anion. Over the years, six human homologs of the catalytic subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the NOX2/gp91phox component present in the phagocyte NADPH oxidase assembly itself, the homologs are now referred to as the NOX family of NADPH oxidases. NOX are complex multidomain proteins with varying requirements for assembly with combinations of other proteins for activity. The recent structural insights acquired on both prokaryotic and eukaryotic NOX open new perspectives for the understanding of the molecular mechanisms inherent to NOX regulation and ROS production (superoxide or hydrogen peroxide). This new structural information will certainly inform new investigations of human disease. As specialized ROS producers, NOX enzymes participate in numerous crucial physiological processes, including host defense, the post-translational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. These diversities of physiological context will be discussed in this review. We also discuss NOX misregulation, which can contribute to a wide range of severe pathologies, such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, or neurodegenerative diseases, giving this family of membrane proteins a strong therapeutic interest.
Collapse
Affiliation(s)
- Annelise Vermot
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Isabelle Petit-Härtlein
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Susan M. E. Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA;
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| |
Collapse
|
17
|
Oxidative Stress in Cancer Cell Metabolism. Antioxidants (Basel) 2021; 10:antiox10050642. [PMID: 33922139 PMCID: PMC8143540 DOI: 10.3390/antiox10050642] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [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
|
18
|
Chen X, Wang Y, Tian J, Shao Y, Zhu B, Wang J, Hua Z. Quantitative Chemical Proteomics Reveals Resveratrol Inhibition of A549 Cell Migration Through Binding Multiple Targets to Regulate Cytoskeletal Remodeling and Suppress EMT. Front Pharmacol 2021; 12:636213. [PMID: 33867987 PMCID: PMC8044895 DOI: 10.3389/fphar.2021.636213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/10/2021] [Indexed: 12/03/2022] Open
Abstract
Resveratrol (RSV), a health-promoting natural product, has been shown to affect various cellular processes in tumor cells. However, the specific protein targets of RSV and the mechanism of action (MOA) of its anticancer effect remain elusive. In this study, the pharmacological activity of RSV was first evaluated in A549 cells, and the results showed that RSV significantly inhibited A549 cell migration but did not affect cell viability. To elucidate the underlying mechanism, a quantitative chemical proteomics approach was employed to identify the protein targets of RSV. A total of 38 target proteins were identified, and proteomic analysis showed that the targets were mainly involved in cytoskeletal remodeling and EMT, which were verified by subsequent in vitro and in vivo assays. In conclusion, RSV inhibits A549 cell migration by binding to multiple targets to regulate cytoskeletal remodeling and suppress EMT.
Collapse
Affiliation(s)
- Xiao Chen
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Yutong Wang
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Tian
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yurou Shao
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Zhu
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zichun Hua
- School of Medicine and Holistic Integrative Medicine and College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,School of Biopharmacy, China Pharmaceutical University, Nanjing, China.,The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
19
|
Ma WF, Boudreau HE, Leto TL. Pan-Cancer Analysis Shows TP53 Mutations Modulate the Association of NOX4 with Genetic Programs of Cancer Progression and Clinical Outcome. Antioxidants (Basel) 2021; 10:antiox10020235. [PMID: 33557266 PMCID: PMC7915715 DOI: 10.3390/antiox10020235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/28/2022] Open
Abstract
Previously, we have shown TGF-β-induced NOX4 expression is involved in the epithelial-to-mesenchymal transition (EMT), a process critical for cancer metastasis, and that wild-type (WT) and mutant (Mut) p53 have divergent effects on TGF-β induction of NOX4: WT-p53 suppresses whereas Mut-p53 augments NOX4 mRNA and protein production in several tumor cell models. We sought to validate and extend our model by analyzing whole-exome data of primary tumor samples in The Cancer Genome Atlas (TCGA). We constructed a Pan-Cancer dataset from 23 tumor types and explored NOX4 expression patterns in relation to EMT and patient survival. NOX4 mRNA levels increase as a function of cancer progression in several cancers and correlate with Mut-p53 mRNA and genes involved in programs of EMT, cellular adhesion, migration, and angiogenesis. Tumor macrophages appear to be a source of NOX2, whose association with genetic programs of cancer progression emulate that of NOX4. Notably, increased NOX4 expression is linked to poorer survival in patients with Mut-TP53, but better survival in patients with WT-TP53. NOX4 is negatively associated with markers of apoptosis and positively with markers of proliferation in patients with Mut-TP53, consistent with their poorer survival. These findings suggest that TP53 mutations could “switch” NOX4 from being protective and an indicator of good prognosis to deleterious by promoting programs favoring cancer progression.
Collapse
|
20
|
Schnelle M, Sawyer I, Anilkumar N, Mohamed BA, Richards DA, Toischer K, Zhang M, Catibog N, Sawyer G, Mongue-Din H, Schröder K, Hasenfuss G, Shah AM. NADPH oxidase-4 promotes eccentric cardiac hypertrophy in response to volume overload. Cardiovasc Res 2021; 117:178-187. [PMID: 31821410 PMCID: PMC7797217 DOI: 10.1093/cvr/cvz331] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 12/07/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Chronic pressure or volume overload induce concentric vs. eccentric left ventricular (LV) remodelling, respectively. Previous studies suggest that distinct signalling pathways are involved in these responses. NADPH oxidase-4 (Nox4) is a reactive oxygen species-generating enzyme that can limit detrimental cardiac remodelling in response to pressure overload. This study aimed to assess its role in volume overload-induced remodelling. METHODS AND RESULTS We compared the responses to creation of an aortocaval fistula (Shunt) to induce volume overload in Nox4-null mice (Nox4-/-) vs. wild-type (WT) littermates. Induction of Shunt resulted in a significant increase in cardiac Nox4 mRNA and protein levels in WT mice as compared to Sham controls. Nox4-/- mice developed less eccentric LV remodelling than WT mice (echocardiographic relative wall thickness: 0.30 vs. 0.27, P < 0.05), with less LV hypertrophy at organ level (increase in LV weight/tibia length ratio of 25% vs. 43%, P < 0.01) and cellular level (cardiomyocyte cross-sectional area: 323 µm2 vs. 379 μm2, P < 0.01). LV ejection fraction, foetal gene expression, interstitial fibrosis, myocardial capillary density, and levels of myocyte apoptosis after Shunt were similar in the two genotypes. Myocardial phospho-Akt levels were increased after induction of Shunt in WT mice, whereas levels decreased in Nox4-/- mice (+29% vs. -21%, P < 0.05), associated with a higher level of phosphorylation of the S6 ribosomal protein (S6) and the eIF4E-binding protein 1 (4E-BP1) in WT compared to Nox4-/- mice. We identified that Akt activation in cardiac cells is augmented by Nox4 via a Src kinase-dependent inactivation of protein phosphatase 2A. CONCLUSION Endogenous Nox4 is required for the full development of eccentric cardiac hypertrophy and remodelling during chronic volume overload. Nox4-dependent activation of Akt and its downstream targets S6 and 4E-BP1 may be involved in this effect.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Arteriovenous Shunt, Surgical
- Cell Cycle Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NADPH Oxidase 2/genetics
- NADPH Oxidase 2/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Phosphorylation
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Ribosomal Protein S6/metabolism
- Signal Transduction
- Ventricular Function, Left
- Ventricular Remodeling
- src-Family Kinases/metabolism
- Mice
Collapse
Affiliation(s)
- Moritz Schnelle
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Iain Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Narayana Anilkumar
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Daniel A Richards
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Min Zhang
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Norman Catibog
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Greta Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Héloïse Mongue-Din
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Ajay M Shah
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
21
|
NOX2-Derived Reactive Oxygen Species in Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7095902. [PMID: 33312338 PMCID: PMC7721506 DOI: 10.1155/2020/7095902] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
The formation of reactive oxygen species (ROS) by the myeloid cell NADPH oxidase NOX2 is critical for the destruction of engulfed microorganisms. However, recent studies imply that ROS, formed by NOX2+ myeloid cells in the malignant microenvironment, exert multiple actions of relevance to the growth and spread of neoplastic cells. By generating ROS, tumor-infiltrating myeloid cells and NOX2+ leukemic myeloid cells may thus (i) compromise the function and viability of adjacent cytotoxic lymphocytes, including natural killer (NK) cells and T cells, (ii) oxidize DNA to trigger cancer-promoting somatic mutations, and (iii) affect the redox balance in cancer cells to control their proliferation and survival. Here, we discuss the impact of NOX2-derived ROS for tumorigenesis, tumor progression, regulation of antitumor immunity, and metastasis. We propose that NOX2 may be a targetable immune checkpoint in cancer.
Collapse
|
22
|
Synergic Crosstalk between Inflammation, Oxidative Stress, and Genomic Alterations in BCR-ABL-Negative Myeloproliferative Neoplasm. Antioxidants (Basel) 2020; 9:antiox9111037. [PMID: 33114087 PMCID: PMC7690801 DOI: 10.3390/antiox9111037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/06/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) have recently been revealed to be related to chronic inflammation, oxidative stress, and the accumulation of reactive oxygen species. It has been proposed that MPNs represent a human inflammation model for tumor advancement, in which long-lasting inflammation serves as the driving element from early tumor stage (over polycythemia vera) to the later myelofibrotic cancer stage. It has been theorized that the starting event for acquired stem cell alteration may occur after a chronic inflammation stimulus with consequent myelopoietic drive, producing a genetic stem cell insult. When this occurs, the clone itself constantly produces inflammatory components in the bone marrow; these elements further cause clonal expansion. In BCR-ABL1-negative MPNs, the driver mutations include JAK 2, MPL, and CALR. Transcriptomic studies of hematopoietic stem cells from subjects with driver mutations have demonstrated the upregulation of inflammation-related genes capable of provoking the development of an inflammatory state. The possibility of acting on the inflammatory state as a therapeutic approach in MPNs appears promising, in which an intervention operating on the pathways that control the synthesis of cytokines and oxidative stress could be effective in reducing the possibility of leukemic progression and onset of complications.
Collapse
|
23
|
NOX4 overexpression is a poor prognostic factor in patients undergoing curative esophagectomy for esophageal squamous cell carcinoma. Surgery 2019; 167:620-627. [PMID: 31889545 DOI: 10.1016/j.surg.2019.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/09/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nox4 has been associated with tumor progression in various types of malignancies. This study aimed to evaluate the importance of the expression of Nox4 in patients undergoing curative esophagectomy for esophageal squamous cell carcinoma. METHODS We reviewed retrospectively 121 patients with esophageal squamous cell carcinoma who had undergone a curative esophagectomy, including 67 patients with overexpression and 54 patients with low expression of Nox4 as evaluated by immunohistochemical analysis. In addition, 2 esophageal squamous cell carcinoma cell lines, TE11 and KYSE270, were treated with the Nox4 inhibitor GKT-137831 to explore the expression of Nox4, cell proliferative activity, and selected downstream pathways in these esophageal squamous cell carcinoma cell lines by Western blot analysis. RESULTS Univariate analysis showed that T1-2 status, absence of nodal metastasis, and low Nox4 expression were associated with greater disease-free survival (P = .001) and overall survival (P < .001), and Nox4 overexpression was an independent prognostic factor of worse disease-free survival and overall survival (P = .013 and P = .007, respectively). The esophageal squamous cell carcinoma cell lines treated with the Nox4 inhibitor GKT-137831 showed decreased cell proliferation in a dose-dependent manner (P < .01). Western blot analysis demonstrated that expression of AKT, phosphorylated AKT, mammalian target of rapamycin, and phosphorylated mammalian target of rapamycin were less in esophageal squamous cell carcinoma cells treated with the Nox4 inhibitor (P < .01). CONCLUSION This study suggests that Nox4 overexpression is a poor prognostic factor for patients with esophageal squamous cell carcinoma undergoing curative esophagectomy.
Collapse
|
24
|
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: 8.2] [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
|
25
|
Mroueh FM, Noureldein M, Zeidan YH, Boutary S, Irani SAM, Eid S, Haddad M, Barakat R, Harb F, Costantine J, Kanj R, Sauleau EA, Ouhtit A, Azar ST, Eid AH, Eid AA. Unmasking the interplay between mTOR and Nox4: novel insights into the mechanism connecting diabetes and cancer. FASEB J 2019; 33:14051-14066. [DOI: 10.1096/fj.201900396rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Fatima Mohsen Mroueh
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mohamed Noureldein
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Youssef H. Zeidan
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- Department of Radiation Oncology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Suzan Boutary
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sara Abou Merhi Irani
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Stéphanie Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mary Haddad
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Rasha Barakat
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frederic Harb
- Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Joseph Costantine
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut, Lebanon
| | - Rouwaida Kanj
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut, Lebanon
| | - Erik-André Sauleau
- Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Sami T. Azar
- Department of Internal Medicine, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, Faculty of Medicine and Medical Center American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, Faculty of Medicine and Medical Center American University of Beirut, Beirut, Lebanon
| |
Collapse
|
26
|
Eun HS, Chun K, Song IS, Oh CH, Seong IO, Yeo MK, Kim KH. High nuclear NADPH oxidase 4 expression levels are correlated with cancer development and poor prognosis in hepatocellular carcinoma. Pathology 2019; 51:579-585. [PMID: 31443922 DOI: 10.1016/j.pathol.2019.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/29/2019] [Accepted: 05/07/2019] [Indexed: 11/30/2022]
Abstract
NADPH oxidase (NOX) is a key source of reactive oxygen species (ROS). This study aimed to verify NOX2 and NOX4 expression levels in hepatocellular carcinoma (HCC). A total of 134 matched pairs of HCC cells and non-tumour hepatocytes from 134 patients were examined by immunohistochemical staining, and the association of NOX2 and NOX4 expression with clinicopathological parameters was analysed. Western blotting in four HCC cell lines and reverse transcription digital droplet polymerase chain reaction (RT-ddPCR) in 20 pairs of HCC and non-tumour tissue samples were also performed to detect NOX4. Cytoplasmic NOX2 and nuclear NOX4 expression levels were shown by immunohistochemistry to be higher in HCC cells than in non-tumour hepatocytes (p<0.001 each). The western blotting results for NOX4 in four HCC cell lines were consistent with the immunohistochemical results. Increased cytoplasmic expression of NOX2 and NOX4 in HCC cells was significantly correlated with liver cirrhosis (p<0.001 and p<0.031, respectively). However, decreased cytoplasmic expression of NOX2 and NOX4 was significantly correlated with advanced pathological TNM stage (p<0.029 and p<0.007, respectively). Multivariate analysis with clinicopathological parameters showed that high nuclear and low cytoplasmic NOX4 expression levels are correlated with short overall survival (p=0 .021). Our findings imply that cytoplasmic NOX2 and nuclear NOX4 expression is upregulated during HCC development. In particular, NOX4 translocation into the nucleus may affect the development and progression of HCC. NOX2 and NOX4 could be diagnostic markers and have therapeutic implications in HCC.
Collapse
Affiliation(s)
- Hyuk Soo Eun
- Department of Internal Medicine, Chungnam National University Hospital, Jung-gu, Daejeon, Republic of Korea; Department of Internal Medicine, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - Kwangsik Chun
- Department of Surgery, Chungnam National University Hospital, Jung-gu, Daejeon, Republic of Korea; Department of Surgery, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - In-Sang Song
- Department of Surgery, Chungnam National University Hospital, Jung-gu, Daejeon, Republic of Korea; Department of Surgery, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - Cheong-Hae Oh
- Department of Internal Medicine, Chungnam National University Hospital, Jung-gu, Daejeon, Republic of Korea; Department of Internal Medicine, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - In-Ock Seong
- Department of Pathology, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea
| | - Kyung-Hee Kim
- Department of Pathology, Chungnam National University School of Medicine, Jung-gu, Daejeon, Republic of Korea.
| |
Collapse
|
27
|
Vakili Saatloo M, Aghbali AA, Koohsoltani M, Yari Khosroushahi A. Akt1 and Jak1 siRNA based silencing effects on the proliferation and apoptosis in head and neck squamous cell carcinoma. Gene 2019; 714:143997. [PMID: 31348981 DOI: 10.1016/j.gene.2019.143997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 01/14/2023]
Abstract
Based on Akt1 and Jak1 key roles in apoptosis and proliferation of many cancers, the aim of this study was to find a new gene therapy strategy by silencing of these main anti-apoptotic genes for HNSCC treatment. Cancerous HN5 and normal HUVEC cell lines were treated with Akt1 and Jak1 siRNAs alone or with each other combined with/without cisplatin. The MTS, flow cytometry, 4',6-diamidino-2-phenylindole staining, real-time PCR and ELISA methods were utilized in this study. The highest percentage of apoptosis was observed in the treatment of Jak1 siRNA/cisplatin group in cancerous HN5 cells (96.5%) where this treatment showed 12.84% apoptosis in normal HUVEC cell line. Cell viability reduced significantly to 64.57% after treatment with Akt1 siRNA in HN5 treated group. Knocking down Akt1 and Jak1 genes using siRNAs could increase levels of apoptosis and reduce proliferation rate in HNSCC indicating the powerful effects of these genes siRNAs with or without chemotherapeutic agents in HNSCC treatment. In conclusion, the combination of siRNA-mediated gene-silencing strategy can be considered as a valuable and safe approach for sensitizing cancer cells to chemotherapeutic agents thus proposed further studies regarding this issue to approve some siRNA based therapeutics for using in clinic.
Collapse
Affiliation(s)
- Maedeh Vakili Saatloo
- Oral and Maxillofacial Department of Pathology, Faculty of Dentistry, Urmia Medical University, Urmia, West Azerbaijan, Iran
| | - Amir Ala Aghbali
- Oral and Maxillofacial Department of Pathology, Faculty of Dentistry, Tabriz Medical University, Tabriz, East Azerbaijan, Iran.
| | - Maryam Koohsoltani
- Oral and Maxillofacial Department of Pathology, Faculty of Dentistry, Tabriz Medical University, Tabriz, East Azerbaijan, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Foo CHJ, Pervaiz S. gRASping the redox lever to modulate cancer cell fate signaling. Redox Biol 2019; 25:101094. [PMID: 30638892 PMCID: PMC6859584 DOI: 10.1016/j.redox.2018.101094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 01/17/2023] Open
Abstract
RAS proteins are critical regulators of signaling networks controlling diverse cellular functions such as cell proliferation and survival and its mutation are among the most powerful oncogenic drivers in human cancers. Despite intense efforts, direct RAS-targeting strategies remain elusive due to its "undruggable" nature. To that end, bulk of the research efforts has been directed towards targeting upstream and/or downstream of RAS signaling. However, the therapeutic efficacies of these treatments are limited in the long run due to the acquired drug resistance in RAS-driven cancers. Interestingly, recent studies have uncovered a potential role of RAS in redox-regulation as well as the interplay between ROS and RAS-associated signaling networks during process of cancer initiation and progression. More specifically, these studies provide ample evidence to implicate RAS as a redox-rheostat, manipulating ROS levels to provide a redox-milieu conducive for carcinogenesis. Importantly, the understanding of RAS-ROS interplay could provide us with novel targetable vulnerabilities for designing therapeutic strategies. In this review, we provide a brief summary of the advances in the field to illustrate the dual role of RAS in redox-regulation and its implications in RAS signaling outcomes and also emerging redox-based strategies to target RAS-driven cancers.
Collapse
Affiliation(s)
- Chuan Han Jonathan Foo
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore
| | - Shazib Pervaiz
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; Medical Science Cluster Cancer Program, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore; National University Cancer Institute, NUHS, Singapore.
| |
Collapse
|
29
|
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen derivatives. Initially, they were considered as metabolic by-products (of mitochondria in particular), which consistently lead to aging and disease. Over the last decades, however, it became increasingly apparent that virtually all eukaryotic cells possess specifically ROS-producing enzymes, namely, NOX NADPH oxidases. In most mammals, there are seven NOX isoforms: three closely related isoforms, NOX1, 2, 3, which are activated by cytoplasmic subunits; NOX4, which appears to be constitutively active; and the EF-hand-containing Ca2+-activated isoforms NOX5 and DUOX1 and 2. Loss-of-function mutations in NOX genes can lead to serious human disease. NOX2 deficiency leads to primary immune deficiency, while DUOX2 deficiency presents as congenital hypothyroidism. Nox-deficient mice provide important tools to explore the physiological functions of various NADPH oxidases as a loss of function in Nox2, Nox3, and Duox2 leads to a spontaneous phenotype. The genetic absence of Nox1, Nox4, and Duox1 does not result in an obvious mouse phenotype (the NOX5 gene is absent in rodents and can therefore not be studied using knockout mice). Since the discovery of the NOX family at the turn of the millennium, much progress in understanding the biochemistry and the physiology of NOX has been made; however many questions remain unanswered to date. This chapter is an overview of our present knowledge on mammalian NOX/DUOX enzymes.
Collapse
Affiliation(s)
- Hélène Buvelot
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Vincent Jaquet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
30
|
Abstract
NOX (NADPH oxidases) are a family of NADPH-dependent transmembrane enzymes that synthesize superoxide and other reactive oxygen species. There are seven isoforms (NOX1-5 and DUOX1-2) which derive from a common ancestral NOX. NOX enzymes are distinguished by different modes of activation, the types of ROS that are produced, the cell types where they are expressed, and distinct functional roles. NOX5 was one of the earliest eukaryotic Nox enzymes to evolve and ironically the last isoform to be discovered in humans. In the time since its discovery, our knowledge of the regulation of NOX5 has expanded tremendously, and we now have a more comprehensive understanding of the molecular mechanisms underlying NOX5-dependent ROS production. In contrast, the cell types where NOX5 is robustly expressed and its functional significance in health and disease remain an underdeveloped area. The goal of this chapter is to provide an up-to-date overview of the mechanisms regulating NOX5 function and its importance in human physiology and pathophysiology.
Collapse
Affiliation(s)
- David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
31
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
32
|
Huo JF, Chen XB. P2X4R silence suppresses glioma cell growth through BDNF/TrkB/ATF4 signaling pathway. J Cell Biochem 2018; 120:6322-6329. [PMID: 30362154 DOI: 10.1002/jcb.27919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023]
Abstract
Purinergic receptor P2X 4 (P2X4R), a member of purinergic channels family and a subtype of ionotropic adenosine triphosphate receptors, plays a critical role in tumorigenesis. Evidence suggested that P2X4R is expressed in rat C6 glioma model, however, its role and the underlying mechanism of action are still unclear in human glioblastoma multiforme (GBM). In the current study, our aim is to examine the function and the molecular basis of P2X4R in GBM. We first observed that GBM cells, U251, T98, U87, U373, and A172 were all high expressed P2X4R, when compared with the normal human astrocytes (NHA) cells. To gain the function of P2X4R, P2X4R silence cells were constructed by transfection with P2X4R small interfering RNA (siRNA). We found that P2X4R deletion impeded T98 and U87 cell viability and proliferation, and further studies indicated that cell apoptosis and caspase-3 activity was increased in T98 and U87 cell transfected with P2X4R siRNA. Subsequently, we confirmed that P2X4R silence suppressed brain-derived neurotrophic factor (BDNF), Trk receptor tyrosine kinases (TrkB), and activating transcription factor 4 (ATF4) expression in T98 and U87 cells. And P2X4R siRNA-induced ATF4-expression inhibition dependent on BDNF/TrkB signaling pathway. The impact of P2X4R silence on T98 and U87 cell growth and apoptosis was reversed by ATF4 overexpression. In summary, this study provides the first evidence that P2X4R plays important roles in GBM cell growth and apoptosis.
Collapse
Affiliation(s)
- Jun-Feng Huo
- Second Ward, Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Xiao-Bing Chen
- Second Ward, Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
33
|
Tang CT, Gao YJ, Ge ZZ. NOX4, a new genetic target for anti-cancer therapy in digestive system cancer. J Dig Dis 2018; 19:578-585. [PMID: 30058122 DOI: 10.1111/1751-2980.12651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/19/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Oxidative stress has been implicated as an important factor in tumorigenesis and tumor progression. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit 4 (NOX4), a substrate of NADPH that can generate H2 O2 reactive oxygen species, has been reported to be highly expressed in gastrointestinal tumors. In this review we summarize the available evidence on the biological function of NOX4 in digestive system tumors by focusing on its correlation with classical cell signaling pathways, including VEGF, MAPK and PI3K/AKT, and with biochemical mediators, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), activator protein (AP)-1 and transforming growth factor (TGF)-β. According to the clinical and database studies on tumors of the digestive system, such as colorectal, gastric and pancreatic cancer, there are significant associations between NOX4 expression and tumor prognosis as well as patient's survival. Animal studies using NOX4 inhibitors such as diphenylene iodonium and GKT137831, which selectively block NOX4, indicate their potential as therapeutic agents for targeting cancer cells.
Collapse
Affiliation(s)
- Chao Tao Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yun Jie Gao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhi Zheng Ge
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
34
|
Zibara K, Zeidan A, Mallah K, Kassem N, Awad A, Mazurier F, Badran B, El-Zein N. Signaling pathways activated by PACAP in MCF-7 breast cancer cells. Cell Signal 2018; 50:37-47. [PMID: 29935235 DOI: 10.1016/j.cellsig.2018.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 01/12/2023]
Abstract
PACAP has opposing roles ranging from activation to inhibition of tumor growth and PACAP agonists/antagonists could be used in tumor therapy. In this study, the effect of PACAP stimulation on signaling pathways was investigated in MCF-7 human adenocarcinoma breast cancer cells. Results showed that MCF-7 cells express VPAC1 and VPAC2, but not PAC1, receptors. In addition, PACAP increased the phosphorylation levels of STAT1, Src and Raf within seconds, confirming their involvement in early stages of PACAP signaling whereas maximal phosphorylation of AKT, ERK and p38 was reached 10 to 20 min later. Moreover, selective inhibition of Src or PI3K resulted in a significant decrease in the phosphorylation of ERK and AKT, but not p38, demonstrating that PACAP signaling follows Src/Raf/ERK and PI3K/AKT pathways. On the other hand, selective inhibition of PLC or PKA resulted in a significant decrease in the phosphorylation of p38, but not AKT or ERK, indicating that PACAP signaling also follows the PLC and PKA/cAMP pathways. Furthermore, PACAP induced ROS through H₂O₂ production whereas pretreatment with NAC inhibitor decreased AKT and ERK phosphorylation, but not p38. Selective NOX2 inhibition affected Src/Raf/Erk and PI3K/Akt pathways, without affecting the p38/PLC/PKA pathway whereas other inhibitors (ML171, VAS2870) had no effect on PACAP induced ROS generation. On the other hand, PACAP induced calcium release, which was decreased by pretreatment with PLC inhibitor. Finally, PACAP stimulation promoted apoptosis by increasing Bax and decreasing Bcl2 expression. In conclusion, we demonstrated that PACAP signaling in MCF-7 cells follows the Src/Raf/ERK and PI3K/AKT pathways and is VPAC1 dependent in a ROS dependent manner, whereas it follows PLC and PKA/cAMP pathways and is VPAC2 dependent through p38 MAP kinase activation involving calcium.
Collapse
Affiliation(s)
- Kazem Zibara
- PRASE, DSST, Lebanese University, Beirut, Lebanon; Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon; Laboratory of Cancer Immunology and Cell Signaling (LCICS), Lebanese University, Faculty of Sciences, Beirut, Lebanon.
| | - Asad Zeidan
- College of Medicine, Department of Basic Medical Sciences, Qatar university, Qatar
| | | | - Nouhad Kassem
- PRASE, DSST, Lebanese University, Beirut, Lebanon; Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ali Awad
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | | | - Bassam Badran
- Laboratory of Cancer Immunology and Cell Signaling (LCICS), Lebanese University, Faculty of Sciences, Beirut, Lebanon
| | - Nabil El-Zein
- PRASE, DSST, Lebanese University, Beirut, Lebanon; Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon; Laboratory of Cancer Immunology and Cell Signaling (LCICS), Lebanese University, Faculty of Sciences, Beirut, Lebanon.
| |
Collapse
|
35
|
Shashni B, Nagasaki Y. Nitroxide radical-containing nanoparticles attenuate tumorigenic potential of triple negative breast cancer. Biomaterials 2018; 178:48-62. [PMID: 29908344 DOI: 10.1016/j.biomaterials.2018.05.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/26/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023]
Abstract
The critical importance of reactive oxygen species (ROS) as oncogene activators and essential secondary messengers in cancer cell survival have been widely reported. Since oxidative stress has been implicated as being pivotal in various cancers, antioxidant therapy seems an apt strategy to abrogate ROS-mediated cellular processes to attenuate cancers. We therefore synthesized ROS scavenging nitroxide radical-containing nanoparticles (RNPs); pH insensitive RNPO and pH sensitive RNPN, to impede the proliferative and metastatic characteristics of the triple negative breast cancer cell line, MDA-MB-231, both in vitro and in vivo. RNPs significantly curtailed the proliferative and clonogenic potential of MDA-MB-231 and MCF-7 cell lines. Inhibition of ROS-mediated migratory and invasive characteristics of MDA-MB-231, via down regulation of NF-κB and MMP-2, was also confirmed. Furthermore, a significant anti-tumor and anti-metastatic potential of RNPs was observed in an MDA-MB-231 mouse xenograft model. Such tumoricidal effects of RNPs were attained with negligible adverse effects, compared to conventional low molecular weight antioxidants, TEMPOL. Thus, the tumoricidal effects of RNPs are suggestive of insights on precedence of nanoparticle-based therapeutics over current low molecular weight antioxidants to curtail ROS-induced tumorigenesis of various cancers.
Collapse
Affiliation(s)
- Babita Shashni
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Center for Research in Isotope and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan.
| |
Collapse
|
36
|
Kitamoto K, Miura Y, Karnan S, Ota A, Konishi H, Hosokawa Y, Sato K. Inhibition of NADPH oxidase 2 induces apoptosis in osteosarcoma: The role of reactive oxygen species in cell proliferation. Oncol Lett 2018; 15:7955-7962. [PMID: 29731909 DOI: 10.3892/ol.2018.8291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/12/2016] [Indexed: 12/31/2022] Open
Abstract
Osteosarcomas (OS) are aggressive tumors that are characterized by dysregulated growth and resistance to apoptosis. Reactive oxygen species (ROS) are thought to be important signal transduction molecules in the regulation of cell growth. ROS-generating nicotinamide adenine dinucleotide phosphate oxidase (NOX) family enzymes have previously been suggested to be involved in neoplastic proliferation. To examine whether NOX-mediated generation of intracellular ROS confers anti-apoptotic activity, and thus a growth advantage, the current study first analyzed the mRNA expression of NOX family members by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in five human OS cell lines. RT-PCR analysis revealed that NOX2 and NOX4 mRNAs were expressed in all the OS cell lines examined, whereas little or no NOX1 and NOX3 mRNAs were detected. By RT-qPCR, NOX2 mRNA expression levels were demonstrated to be higher than NOX4 mRNA expression levels. The viability of OS cells decreased in a dose-dependent manner with treatment of diphenylene iodonium (DPI), an inhibitor of flavoprotein-dependent oxidase. DPI treatment was observed to reduce intracellular ROS levels by ~50%, and increase the frequency of apoptosis by 30%. Notably, small interfering RNAs (siRNAs) targeting NOX2 significantly suppressed ROS generation; ROS depletion by DPI or NOX2 siRNAs induced apoptosis in OS cells. Together, the results of the present study indicate that NOX2-mediated ROS generation promotes cell survival and ROS depletion leads to apoptosis, thus highlighting the NOX2-ROS signaling pathway as a potential therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Kazumasa Kitamoto
- Department of Orthopedic Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Yuji Miura
- Department of Pathophysiology, Aichi Medical University School of Nursing, Nagakute, Aichi 480-1195, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Keiji Sato
- Department of Orthopedic Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
37
|
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: 79] [Impact Index Per Article: 13.2] [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
|
38
|
Lin XL, Yang L, Fu SW, Lin WF, Gao YJ, Chen HY, Ge ZZ. Overexpression of NOX4 predicts poor prognosis and promotes tumor progression in human colorectal cancer. Oncotarget 2018; 8:33586-33600. [PMID: 28422720 PMCID: PMC5464892 DOI: 10.18632/oncotarget.16829] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/25/2017] [Indexed: 12/20/2022] Open
Abstract
NADPH oxidase 4 (NOX4), a major source of reactive oxygen species (ROS) production, has been increasingly reported to be involved in tumorigenesis and/or tumor progression, but limited data are available regarding the role of NOX4 in colorectal carcinoma (CRC). We retrieved six independent investigations from Oncomine database and found that NOX4 is highly expressed in CRC tissues compared with corresponding normal controls. Similar results were also found in clinical specimens at both mRNA and protein levels. Immunohistochemical analysis indicated that NOX4 overexpression was highly correlated with T classification, N classification, distant metastasis, and poor prognosis of CRC patients, which was also confirmed by GSE14333 and GSE17536 datasets from the Gene Expression Omnibus. Furthermore, we demonstrated that when NOX4 expression was knocked down by siRNAs, cell proliferation, cell-cycle and apoptosis, migration and invasion were significantly altered in CRC cell lines HCT116 and LOVO. Meanwhile, NOX4 promoted cancer cell proliferation and apoptosis, migration and invasion by regulating the expression of relevant genes. By these approaches we aim to elucidate NOX4 may be a reliable prognostic factor or therapeutic target in CRC.
Collapse
Affiliation(s)
- Xiao-Lu Lin
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Li Yang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Seng-Wang Fu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Wen-Feng Lin
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Yun-Jie Gao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Hao-Yan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Zhi-Zheng Ge
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200001, China
| |
Collapse
|
39
|
Yu T, Li L, Liu W, Ya B, Cheng H, Xin Q. Silencing of NADPH Oxidase 4 Attenuates Hypoxia Resistance in Neuroblastoma Cells SH-SY5Y by Inhibiting PI3K/Akt-Dependent Glycolysis. Oncol Res 2018; 27:525-532. [PMID: 29426376 PMCID: PMC7848326 DOI: 10.3727/096504018x15179668157803] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-induced chemoresistance is a major obstacle in the development of effective cancer therapy. In our study, the reversal abilities of NADPH oxidase 4 (NOX4) silence on hypoxia resistance and the potential mechanism were investigated. Our data showed that the expression of NOX4 was upregulated in human neuroblastoma cells SH-SY5Y under hypoxia condition time dependently. Knockdown of NOX4 expression by siRNA inhibited glycolysis induced by hypoxia through decreasing the expression of glycolysis-related proteins (HIF-1α, LDHA, and PDK1), decreasing glucose uptake, lactate production, and ROS production, while increasing mitochondria membrane potential. Moreover, NOX4 silence inhibited cell growth under hypoxia condition through suppressing cell proliferation and proliferation-related proteins (Ki-67 and PCNA) compared with the hypoxia 24 h + siRNA NC group. Further, Western blot experiments exhibited that NOX4 siRNA could downregulate the rate of p-Akt/Akt. Treatment with PI3K/Akt signaling activator IGF-1 blocked, while treatment with Akt inhibitor perifosine enhanced the inhibitory effect of si-NOX4 on glycolysis and cell growth. In summary, knockdown of NOX4 had the ability of reversing hypoxia resistance, and the major mechanism is considered to be the inhibition of glycolysis and cell growth via the PI3K/Akt signaling pathway. Therefore, NOX4 could be a novel target against hypoxia resistance in neuroblastoma.
Collapse
Affiliation(s)
- Ting Yu
- Department of Physiology, Jining Medical University, Jining, Shandong, P.R. China
| | - Lei Li
- Department of Diagnosis, Jining Medical University, Jining, Shandong, P.R. China
| | - Wenyan Liu
- Department of Physiology, Jining Medical University, Jining, Shandong, P.R. China
| | - Bailiu Ya
- Department of Physiology, Jining Medical University, Jining, Shandong, P.R. China
| | - Hongju Cheng
- Department of Physiology, Jining Medical University, Jining, Shandong, P.R. China
| | - Qing Xin
- Department of Physiology, Jining Medical University, Jining, Shandong, P.R. China
| |
Collapse
|
40
|
Hegedűs C, Kovács K, Polgár Z, Regdon Z, Szabó É, Robaszkiewicz A, Forman HJ, Martner A, Virág L. Redox control of cancer cell destruction. Redox Biol 2018; 16:59-74. [PMID: 29477046 PMCID: PMC5842284 DOI: 10.1016/j.redox.2018.01.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023] Open
Abstract
Redox regulation has been proposed to control various aspects of carcinogenesis, cancer cell growth, metabolism, migration, invasion, metastasis and cancer vascularization. As cancer has many faces, the role of redox control in different cancers and in the numerous cancer-related processes often point in different directions. In this review, we focus on the redox control mechanisms of tumor cell destruction. The review covers the tumor-intrinsic role of oxidants derived from the reduction of oxygen and nitrogen in the control of tumor cell proliferation as well as the roles of oxidants and antioxidant systems in cancer cell death caused by traditional anticancer weapons (chemotherapeutic agents, radiotherapy, photodynamic therapy). Emphasis is also put on the role of oxidants and redox status in the outcome following interactions between cancer cells, cytotoxic lymphocytes and tumor infiltrating macrophages.
Collapse
Affiliation(s)
- Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Kovács
- MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
| | - Zsuzsanna Polgár
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Szabó
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary.
| |
Collapse
|
41
|
Hong S, Kwon J, Hiep NT, Sim SJ, Kim N, Kim KH, Lee D, Mar W. The isoflavones and extracts from Maclura tricuspidata fruit protect against neuronal cell death in ischemic injury via induction of Nox4-targeting miRNA-25, miRNA-92a, and miRNA-146a. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
42
|
Yamamoto T, Nakano H, Shiomi K, Wanibuchi K, Masui H, Takahashi T, Urano Y, Kamata T. Identification and Characterization of a Novel NADPH Oxidase 1 (Nox1) Inhibitor That Suppresses Proliferation of Colon and Stomach Cancer Cells. Biol Pharm Bull 2017; 41:419-426. [PMID: 29269607 DOI: 10.1248/bpb.b17-00804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) generated by reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox)1 mediate cellular signalings involved in normal physiological processes, and aberrant control of Nox1 has been implicated in the pathogenesis of various diseases. Therefore, Nox1 could have great potential as a therapeutic target. Here, we identified a novel Nox1 inhibitor, NOS31 secreted from Stretomyces sp. and analyzed its chemical structure. Furthermore, NOS31 was found to selectively inhibit Nox1-mediated ROS generation, with only a marginal effect on other Nox isoforms (Nox2-5) and no ROS scavenging activity. This compound blocked both Nox organizer 1 (NOXO1)/Nox activator 1 (NOXA1)-dependent and phorbol 12-myristate 13-acetate-stimulated Nox1-mediated ROS production in colon cancer cells. NOS31 inhibited the proliferation of several colon carcinoma and gastric cancer cell lines that upregulate the Nox1 system, whereas it had no appreciable effect on normal cells with low levels of Nox1. The finding suggests that NOS31 is a unique, potent Nox1 inhibitor of microbial origin and raises its possibility as a therapeutic agent for inhibiting gastrointestinal cancer cell growth.
Collapse
Affiliation(s)
| | | | - Kazuro Shiomi
- Kitasato Institute for Life Sciences, Kitasato University.,Graduate School of Infection Control Sciences, Kitasato University
| | | | - Hisashi Masui
- Faculty of Pharmaceutical Sciences, Yokohama College of Pharmacy
| | | | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo.,Graduate School of Medicine, The University of Tokyo
| | - Tohru Kamata
- Department of Molecular Biology and Biochemistry, Shinshu University Graduate School of Medicine
| |
Collapse
|
43
|
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: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
44
|
Hu L, Zhang X, Wang J, Wang S, Amin HM, Shi P. Involvement of oncogenic tyrosine kinase NPM-ALK in trifluoperazine-induced cell cycle arrest and apoptosis in ALK+ anaplastic large cell lymphoma. Hematology 2017; 23:284-290. [DOI: 10.1080/10245332.2017.1396045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Linlin Hu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
- Key Laboratory of Organofluorine Chemistry, Chinese Academy of Sciences, Shanghai Institute of Organic Chemistry, Shanghai, People’s Republic of China
| | - Xiaonan Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Jian Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Song Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Hesham M. Amin
- Department of Hematopathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People’s Republic of China
- Key Laboratory of Organofluorine Chemistry, Chinese Academy of Sciences, Shanghai Institute of Organic Chemistry, Shanghai, People’s Republic of China
| |
Collapse
|
45
|
The emerging role of NADPH oxidase NOX5 in vascular disease. Clin Sci (Lond) 2017; 131:981-990. [PMID: 28473473 DOI: 10.1042/cs20160846] [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: 02/22/2017] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 01/11/2023]
Abstract
Oxidative stress is a consequence of up-regulation of pro-oxidant enzyme-induced reactive oxygen species (ROS) production and concomitant depletion of antioxidants. Elevated levels of ROS act as an intermediate and are the common denominator for various diseases including diabetes-associated macro-/micro-vascular complications and hypertension. A range of enzymes are capable of generating ROS, but the pro-oxidant enzyme family, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs), are the only enzymes known to be solely dedicated to ROS generation in the vascular tissues, kidney, aortas and eyes. While there is convincing evidence for a role of NOX1 in vascular and eye disease and for NOX4 in renal injury, the role of NOX5 in disease is less clear. Although NOX5 is highly up-regulated in humans in disease, it is absent in rodents. Thus, so far it has not been possible to study NOX5 in traditional mouse or rat models of disease. In the present review, we summarize and critically analyse the emerging evidence for a pathophysiological role of NOX5 in disease including the expression, regulation and molecular and cellular mechanisms which have been demonstrated to be involved in NOX5 activation.
Collapse
|
46
|
The Role of NOX4 and TRX2 in Angiogenesis and Their Potential Cross-Talk. Antioxidants (Basel) 2017; 6:antiox6020042. [PMID: 28594389 PMCID: PMC5488022 DOI: 10.3390/antiox6020042] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) family is the major source of reactive oxygen species (ROS) in the vascular system. In this family, NOX4, a constitutive active form of NOXs, plays an important role in angiogenesis. Thioredoxin 2 (TRX2) is a key mitochondrial redox protein that maintains normal protein function and also provides electrons to peroxiredoxin 3 (PRX3) to scavenge H₂O₂ in mitochondria. Angiogenesis, a process of new blood vessel formation, is involved in a variety of physiological processes and pathological conditions. It seems to be paradoxical for ROS-producing NOX4 and ROS-scavenging TRX2 to have a similar role in promoting angiogenesis. In this review, we will focus on data supporting the role of NOX4 and TRX2 in angiogenesis and their cross-talks and discuss how ROS can positively or negatively regulate angiogenesis, depending on their species, levels and locations. NOX4 and TRX2-mediated ROS signaling could be promising targets for the treatment of angiogenesis-related diseases.
Collapse
|
47
|
Moloney JN, Cotter TG. ROS signalling in the biology of cancer. Semin Cell Dev Biol 2017; 80:50-64. [PMID: 28587975 DOI: 10.1016/j.semcdb.2017.05.023] [Citation(s) in RCA: 1173] [Impact Index Per Article: 167.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/17/2017] [Accepted: 05/29/2017] [Indexed: 12/19/2022]
Abstract
Increased reactive oxygen species (ROS) production has been detected in various cancers and has been shown to have several roles, for example, they can activate pro-tumourigenic signalling, enhance cell survival and proliferation, and drive DNA damage and genetic instability. Counterintuitively ROS can also promote anti-tumourigenic signalling, initiating oxidative stress-induced tumour cell death. Tumour cells express elevated levels of antioxidant proteins to detoxify elevated ROS levels, establish a redox balance, while maintaining pro-tumourigenic signalling and resistance to apoptosis. Tumour cells have an altered redox balance to that of their normal counterparts and this identifies ROS manipulation as a potential target for cancer therapies. This review discusses the generation and sources of ROS within tumour cells, the regulation of ROS by antioxidant defence systems, as well as the effect of elevated ROS production on their signalling targets in cancer. It also provides an insight into how pro- and anti-tumourigenic ROS signalling pathways could be manipulated in the treatment of cancer.
Collapse
Affiliation(s)
- Jennifer N Moloney
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Thomas G Cotter
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland.
| |
Collapse
|
48
|
Nishida T, Hattori K, Watanabe K. The regulatory and signaling mechanisms of the ASK family. Adv Biol Regul 2017; 66:2-22. [PMID: 28669716 DOI: 10.1016/j.jbior.2017.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 01/05/2023]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) was identified as a MAP3K that activates the JNK and p38 pathways, and subsequent studies have reported ASK2 and ASK3 as members of the ASK family. The ASK family is activated by various intrinsic and extrinsic stresses, including oxidative stress, ER stress and osmotic stress. Numerous lines of evidence have revealed that members of the ASK family are critical for signal transduction systems to control a wide range of stress responses such as cell death, differentiation and cytokine induction. In this review, we focus on the precise signaling mechanisms of the ASK family in response to diverse stressors.
Collapse
Affiliation(s)
- Takuto Nishida
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Kazuki Hattori
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| |
Collapse
|
49
|
NOX2, NOX4, and mitochondrial-derived reactive oxygen species contribute to angiopoietin-1 signaling and angiogenic responses in endothelial cells. Vascul Pharmacol 2017; 92:22-32. [DOI: 10.1016/j.vph.2017.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 03/13/2017] [Accepted: 03/18/2017] [Indexed: 11/17/2022]
|
50
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017; 104:144-164. [PMID: 28088622 DOI: 10.1016/j.freeradbiomed.2017.01.004] [Citation(s) in RCA: 626] [Impact Index Per Article: 89.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS), a group of highly reactive ions and molecules, are increasingly being appreciated as powerful signaling molecules involved in the regulation of a variety of biological processes. Indeed, their role is continuously being delineated in a variety of pathophysiological conditions. For instance, cancer cells are shown to have increased ROS levels in comparison to their normal counterparts. This is partly due to an enhanced metabolism and mitochondrial dysfunction in cancer cells. The escalated ROS generation in cancer cells contributes to the biochemical and molecular changes necessary for the tumor initiation, promotion and progression, as well as, tumor resistance to chemotherapy. Therefore, increased ROS in cancer cells may provide a unique opportunity to eliminate cancer cells via elevating ROS to highly toxic levels intracellularly, thereby, activating various ROS-induced cell death pathways, or inhibiting cancer cell resistance to chemotherapy. Such results can be achieved by using agents that either increase ROS generation, or inhibit antioxidant defense, or even a combination of both. In fact, a large variety of anticancer drugs, and some of those currently under clinical trials, effectively kill cancer cells and overcome drug resistance via enhancing ROS generation and/or impeding the antioxidant defense mechanism. This review focuses on our current understanding of the tumor promoting (tumorigenesis, angiogenesis, invasion and metastasis, and chemoresistance) and the tumor suppressive (apoptosis, autophagy, and necroptosis) functions of ROS, and highlights the potential mechanism(s) involved. It also sheds light on a very novel and an actively growing field of ROS-dependent cell death mechanism referred to as ferroptosis.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE; Al Jalila Foundation Research Centre, P.O. Box 300100, Dubai, UAE.
| | - Anees Rahman
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|