1
|
Zhou Y, Wang M, Qian Y, Yu D, Zhang J, Fu M, Zhang X, Qin R, Ji R, Zhang X, Gu J. PRDX2 promotes gastric cancer progression by forming a feedback loop with PKM2/STAT3 axis. Cell Signal 2025; 127:111586. [PMID: 39761843 DOI: 10.1016/j.cellsig.2024.111586] [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: 07/12/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Peroxiredoxin 2 (PRDX2) is an antioxidant enzyme that has been reported to be overexpressed in various cancers. However, the role of PRDX2 in gastric cancer progression and its underlying mechanism remains unclear. Herein, we revealed the function of PRDX2 in gastric cancer progression and explored its molecule mechanism. We identified that PRDX2 was upregulated and associated with poor prognosis in gastric cancer. The knockdown of PRDX2 inhibited the proliferation, migration and invasion of gastric cancer cells in vitro and suppressed tumor growth in vivo. Mechanistically, PRDX2 interacted with PKM2 (pyruvate kinase isozyme type M2) and protected PKM2 from ubiquitination and degradation, which enhanced glycolysis in gastric cancer cells. The interaction between PRDX2 and PKM2 also enhanced the binding affinity between PKM2 and importin α5, which induced PKM2 nuclear translocation and activated STAT3 signaling pathway. In addition, STAT3 (signal transducer and activator of transcription 3) was identified to bind to PRDX2 gene promoter and upregulate PRDX2 expression, which forms a positive regulatory feedback loop in gastric cancer cells. The present study unravels the biological role of PRDX2 in cancer progression and illustrates the underlying molecular mechanism, which may provide a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Kunshan Biomedical Big Data Innovation Application Laboratory, Kunshan Hospital Affiliated to Jiangsu University /Kunshan First People's Hospital, Kunshan 215300, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Min Fu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Rong Qin
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Runbi Ji
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Kunshan Biomedical Big Data Innovation Application Laboratory, Kunshan Hospital Affiliated to Jiangsu University /Kunshan First People's Hospital, Kunshan 215300, China.
| | - Jianmei Gu
- Department of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong 226300, China.
| |
Collapse
|
2
|
Guo M, Guo D, Liao L, Zhang X, Wang Z, Zhou Q, Chen P, Li R, Han B, Bao G, Zhang B. Ethanolic extract from Sophora moorcroftiana inhibit cell proliferation and alter the mechanical properties of human cervical cancer. BMC Complement Med Ther 2024; 24:212. [PMID: 38831394 PMCID: PMC11149180 DOI: 10.1186/s12906-024-04502-5] [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: 11/25/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Cervical cancer is one of the most common gynecological malignancies. Previous studies have shown that the ethanol extract of Sophora moorcroftiana seeds (EESMS) possesses an antiproliferative effect on several tumors in vitro. Therefore, in this study, we assessed the impact of EESMS on human cervical carcinoma (HeLa) cell proliferation. METHODS The proliferation and apoptotic effects of HeLa cells treated with EESMS were evaluated using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay, dual acridine orange/ethidium bromide double staining, flow cytometry, and western blotting. Single-cell level atomic force microscopy (AFM) was conducted to detect the mechanical properties of HeLa cells, and proteomics and bioinformatics methods were used to elucidate the molecular mechanisms of EESMS. RESULTS EESMS treatment inhibited HeLa cell proliferation by blocking the G0/G1 phase, increasing the expression of Caspase-3 and affecting its mechanical properties, and the EESMS indicated no significant inhibitory effect on mouse fibroblasts L929 cell line. In total, 218 differentially expressed proteins were identified using two-dimensional electrophoresis, and eight differentially expressed proteins were successfully identified using matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry. The differentially expressed proteins were involved in various cellular and biological processes. CONCLUSION This study provides a perspective on how cells change through biomechanics and a further theoretical foundation for the future application of Sophora moorcroftiana as a novel low-toxicity chemotherapy medication for treating human cervical cancer.
Collapse
Affiliation(s)
- Manli Guo
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Northwest new village No.1, Lanzhou, 730030, PR China
| | - Dingcheng Guo
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Lingzi Liao
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Xiao Zhang
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Zhilong Wang
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Qiaozhen Zhou
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Ping Chen
- Chengdu Stomatological Hospital, NO. 17, South Section of Chunxi Road, Jinjiang District, Chengdu, 610020, PR China
| | - Ruiping Li
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China
- Gansu Province Key Lab of Maxillofacial Reconstruction and Intelligent Manufacturing, Donggang West Road 199, Lanzhou, 730000, PR China
| | - Bing Han
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Northwest new village No.1, Lanzhou, 730030, PR China
| | - Guangjie Bao
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Northwest new village No.1, Lanzhou, 730030, PR China.
| | - Baoping Zhang
- School (Hospital) of Stomatology, Lanzhou University, Donggang West Road 199, Lanzhou, 730000, PR China.
- Gansu Province Key Lab of Maxillofacial Reconstruction and Intelligent Manufacturing, Donggang West Road 199, Lanzhou, 730000, PR China.
| |
Collapse
|
3
|
Wang TH, Shen YW, Chen HY, Chen CC, Lin NC, Shih YH, Hsia SM, Chiu KC, Shieh TM. Arecoline Induces ROS Accumulation, Transcription of Proinflammatory Factors, and Expression of KRT6 in Oral Epithelial Cells. Biomedicines 2024; 12:412. [PMID: 38398015 PMCID: PMC10887121 DOI: 10.3390/biomedicines12020412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Areca nut is a major contributor to the high prevalence of oral cancer in Asia. The precise mechanisms by which areca nut stimulates mucosal cells and contributes to the progression of oral cancer urgently require clarification. The current study aimed to assess the effects of arecoline on the normal human gingival epithelium cell line S-G. Cell viability, levels of reactive oxygen species (ROS), protein expression, cellular morphology, and gene expression were evaluated using the MTT test, flow cytometry, Western blot analysis, optical or confocal microscopy, and RT-qPCR. Keratin (KRT6) analysis involved matched normal and cancer tissues from clinical head and neck specimens. The results demonstrated that 12.5 µg/mL of arecoline induced ROS production, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) mRNA expression in S-G cells. This activation of the MAPK/ERK pathway increased KRT6 expression while limiting cell migration. In head and neck cancer tissues, KRT6B gene expression exceeded that of normal tissues. This study confirms that arecoline induces ROS accumulation in normal cells, leading to the secretion of proinflammatory factors and KRT6 expression. This impedes oral mucosal healing, thereby promoting the progression of oral cancer.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Biobank, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yen-Wen Shen
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| | - Hsin-Ying Chen
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| | - Chih-Chieh Chen
- Department of Sports Medicine, China Medical University, Taichung 404328, Taiwan
| | - Nan-Chin Lin
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, Changhua 505029, Taiwan
- Department of Oral and Maxillofacial Surgery, Changhua Christian Hospital, Changhua 500011, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Kuo-Chou Chiu
- Division of General Dentistry, Taichung Armed Forces General Hospital, Taichung 411228, Taiwan
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
4
|
Liu Y, Wang P, Hu W, Chen D. New insights into the roles of peroxiredoxins in cancer. Biomed Pharmacother 2023; 164:114896. [PMID: 37210897 DOI: 10.1016/j.biopha.2023.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023] Open
Abstract
Oxidative stress is one of the hallmarks of cancer. Tumorigenesis and progression are accompanied by elevated reactive oxygen species (ROS) levels and adaptive elevation of antioxidant expression levels. Peroxiredoxins (PRDXs) are among the most important antioxidants and are widely distributed in a variety of cancers. PRDXs are involved in the regulation of a variety of tumor cell phenotypes, such as invasion, migration, epithelial-mesenchymal transition (EMT) and stemness. PRDXs are also associated with tumor cell resistance to cell death, such as apoptosis and ferroptosis. In addition, PRDXs are involved in the transduction of hypoxic signals in the TME and in the regulation of the function of other cellular components of the TME, such as cancer-associated fibroblasts (CAFs), natural killer (NK) cells and macrophages. This implies that PRDXs are promising targets for cancer treatment. Of course, further studies are needed to realize the clinical application of targeting PRDXs. In this review, we highlight the role of PRDXs in cancer, summarizing the basic features of PRDXs, their association with tumorigenesis, their expression and function in cancer, and their relationship with cancer therapeutic resistance.
Collapse
Affiliation(s)
- Yan Liu
- First Department of Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Pu Wang
- Department of Emergency, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Weina Hu
- Department of General Practice, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China.
| | - Da Chen
- Department of Emergency, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China.
| |
Collapse
|
5
|
Khan SU, Fatima K, Aisha S, Hamza B, Malik F. Redox balance and autophagy regulation in cancer progression and their therapeutic perspective. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:12. [PMID: 36352310 DOI: 10.1007/s12032-022-01871-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022]
Abstract
Cellular ROS production participates in various cellular functions but its accumulation decides the cell fate. Malignant cells have higher levels of ROS and active antioxidant machinery, a characteristic hallmark of cancer with an outcome of activation of stress-induced pathways like autophagy. Autophagy is an intracellular catabolic process that produces alternative raw materials to meet the energy demand of cells and is influenced by the cellular redox state thus playing a definite role in cancer cell fate. Since damaged mitochondria are the main source of ROS in the cell, however, cancer cells remove them by upregulating the process of mitophagy which is known to play a decisive role in tumorigenesis and tumor progression. Chemotherapy exploits cell machinery which results in the accumulation of toxic levels of ROS in cells resulting in cell death by activating either of the pathways like apoptosis, necrosis, ferroptosis or autophagy in them. So understanding these redox and autophagy regulations offers a promising method to design and develop new cancer therapies that can be very effective and durable for years. This review will give a summary of the current therapeutic molecules targeting redox regulation and autophagy for the treatment of cancer. Further, it will highlight various challenges in developing anticancer agents due to autophagy and ROS regulation in the cell and insights into the development of future therapies.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
| | - Baseerat Hamza
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India.
| |
Collapse
|
6
|
Xu D, Zhu X, Ren J, Huang S, Xiao Z, Jiang H, Tan Y. Quantitative proteomic analysis of cervical cancer based on TMT-labeled quantitative proteomics. J Proteomics 2022; 252:104453. [PMID: 34915198 DOI: 10.1016/j.jprot.2021.104453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/18/2022]
Abstract
Cervical cancer is the second most common gynecological malignancy, which immensely threatens the well-being of women. However, the pathogenesis of cervical cancer is still unclear. Using tandem mass tags-labeled quantitative proteomic technology and bioinformatics tools, we analyzed the exfoliated cervical cells from the normal and cervical cancer groups to establish a cancer-specific protein profile, thereby identifying key proteins related to cervical oncogenesis. When compared with the normal group, a total of 351 differentially expressed proteins were identified in the cervical cancer group, including 247 up-regulated and 104 down-regulated proteins. Gene ontology function annotation revealed that the differentially expressed proteins were mainly involved in the single-multicellular organism process, multicellular organismal process, and negative regulation of biological process. These proteins were discerned to play a role in the extracellular membrane-bounded organelle, exosome of cell components, protein binding, structural molecule activity, and enzyme binding of molecular functions. The results of Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment proved that these differentially expressed proteins were mainly involved in PI3K - Akt, ECM-receptor interaction, complement and coagulation cascades, and other signaling pathways. Particularly, peroxiredoxin-2 may be involved in cervical tumor oncogenesis through inhibition of apoptosis signaling. SIGNIFICANCE: In this study, we determined that the proteins of the cervical cancer group exhibited qualitative and quantitative changes, and a total of 351 differentially expressed proteins were identified. The functions and signaling pathways of these differentially expressed proteins have laid a theoretical foundation for elucidating the molecular mechanism of cervical cancer.
Collapse
Affiliation(s)
- Dianqin Xu
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Xiaoyu Zhu
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Ji Ren
- School of Laboratory Medicine, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Shan Huang
- School of Laboratory Medicine, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Ziwen Xiao
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Hongmei Jiang
- School of Laboratory Medicine, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Yujie Tan
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, China; School of Laboratory Medicine, Guizhou Medical University, Guiyang 550004, Guizhou, China.
| |
Collapse
|
7
|
Vasilogianni AM, Al-Majdoub ZM, Achour B, Peters SA, Rostami-Hodjegan A, Barber J. Proteomics of colorectal cancer liver metastasis: A quantitative focus on drug elimination and pharmacodynamics effects. Br J Clin Pharmacol 2021; 88:1811-1823. [PMID: 34599518 PMCID: PMC9299784 DOI: 10.1111/bcp.15098] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/09/2022] Open
Abstract
Aims This study aims to quantify drug‐metabolising enzymes, transporters, receptor tyrosine kinases (RTKs) and protein markers (involved in pathways affected in cancer) in pooled healthy, histologically normal and matched cancerous liver microsomes from colorectal cancer liver metastasis (CRLM) patients. Methods Microsomal fractionation was performed and pooled microsomes were prepared. Global and accurate mass and retention time liquid chromatography–mass spectrometry proteomics were used to quantify proteins. A QconCAT (KinCAT) for the quantification of RTKs was designed and applied for the first time. Physiologically based pharmacokinetic (PBPK) simulations were performed to assess the contribution of altered abundance of drug‐metabolising enzymes and transporters to changes in pharmacokinetics. Results Most CYPs and UGTs were downregulated in histologically normal relative to healthy samples, and were further reduced in cancer samples (up to 54‐fold). The transporters, MRP2/3, OAT2/7 and OATP2B1/1B3/1B1 were downregulated in CRLM. Application of abundance data in PBPK models for substrates with different attributes indicated substantially lower (up to 13‐fold) drug clearance when using cancer‐specific instead of default parameters in cancer population. Liver function markers were downregulated, while inflammation proteins were upregulated (by up to 76‐fold) in cancer samples. Various pharmacodynamics markers (e.g. RTKs) were altered in CRLM. Using global proteomics, we examined proteins in pathways relevant to cancer (such as metastasis and desmoplasia), including caveolins and collagen chains, and confirmed general over‐expression of such pathways. Conclusion This study highlights impaired drug metabolism, perturbed drug transport and altered abundance of cancer markers in CRLM, demonstrating the importance of population‐specific abundance data in PBPK models for cancer.
Collapse
Affiliation(s)
- Areti-Maria Vasilogianni
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| | | | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK.,Certara Inc (Simcyp Division), Sheffield, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Lee S, Lee JY, Lee EW, Park S, Kang DH, Min C, Lee DJ, Kang D, Song J, Kwon J, Kang SW. Absence of Cytosolic 2-Cys Prx Subtypes I and II Exacerbates TNF-α-Induced Apoptosis via Different Routes. Cell Rep 2020; 26:2194-2211.e6. [PMID: 30784599 DOI: 10.1016/j.celrep.2019.01.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/04/2018] [Accepted: 01/22/2019] [Indexed: 01/19/2023] Open
Abstract
There are abundant peroxiredoxin (Prx) enzymes, but an increase of cellular H2O2 level always happens in apoptotic cells. Here, we show that cellular H2O2 switches different apoptosis pathways depending on which type of Prx enzyme is absent. TNF-α-induced H2O2 burst preferentially activates the DNA damage-dependent apoptosis pathway in the absence of PrxI. By contrast, the same H2O2 burst stimulates the RIPK1-dependent apoptosis pathway in the absence of PrxII by inducing the destruction of cIAP1 in caveolar membrane. Specifically, H2O2 induces the oxidation of Cys308 residue in the cIAP1-BIR3 domain, which induces the dimerization-dependent E3 ligase activation. Thus, the reduction in cIAP level by the absence of PrxII triggers cell-autonomous apoptosis in cancer cells and tumors. Such differential functions of PrxI and PrxII are mediated by interaction with H2AX and cIAP1, respectively. Collectively, this study reveals the distinct switch roles of 2-Cys Prx isoforms in apoptosis signaling.
Collapse
Affiliation(s)
- Sunmi Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Joo Young Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Eun Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Sujin Park
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Dong Hoon Kang
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Chengchun Min
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Doo Jae Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Dongmin Kang
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Jaewhan Song
- Department of Biochemistry, Yonsei University, Seoul 03722, Korea
| | - Jongbum Kwon
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea; Research Center for Cell Homeostasis, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
9
|
Najafi Z, Mohamadnia A, Ahmadi R, Mahmoudi M, Bahrami N, Khosravi A, Jamaati H, Tabarsi P, Kazem Pour Dizaji M, Shirian S. Proteomic and genomic biomarkers for Non-Small Cell Lung Cancer: Peroxiredoxin, Haptoglobin, and Alpha-1 antitrypsin. Cancer Med 2020; 9:3974-3982. [PMID: 32232956 PMCID: PMC7286458 DOI: 10.1002/cam4.3019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/28/2020] [Accepted: 03/08/2020] [Indexed: 12/18/2022] Open
Abstract
Background The development of lung cancer is a multifactorial process that involves the environmental and genetic factors. The mortality rate of this cancer is higher than breast, colorectal, and prostate cancers. In this study, we try to analyze the proteome of patients with Non‐Small Cell Lung Cancer (NSCLC) and compare it with the healthy samples. Methods This study has compared 30 lung tissue samples from patients with NSCLC and 30 healthy samples using proteomics and RT‐PCR. Hence, tissue samples were obtained from the surgical ward in sterile conditions, and then, protein extraction applied to them. At the next stage, two‐dimensional electrophoresis and mass spectrometry LCMS/MS were performed for protein isolation and sequencing, respectively. Results The proteome analysis identified more than 40 differences in proteomic pattern of normal lung tissues compared to lung tissues with NSCLC. Peroxiredoxin, Haptoglobin, and Alpha‐1 antitrypsin proteins were identified. Molecularly, it has also been shown that the two main proteins of Peroxiredoxin‐2 and Alpha‐1 antitrypsin were upregulated, and the expression of Haptoglobin protein was downregulated in cancer tissue. Conclusion The results of this study showed that there are some differences in term of protein content between the normal and cancerous lung tissues. Further studies are needed to evaluate these proteins that investigate whether these proteins can candidate as biomarkers to use in the early diagnosis of patients with NSCLC.
Collapse
Affiliation(s)
- Zahra Najafi
- Department of Biology, Faculty of Basic Sciences, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Abdolreza Mohamadnia
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rahim Ahmadi
- Department of Biology, Faculty of Basic Sciences, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Minoo Mahmoudi
- Department of Biology, Faculty of Basic Sciences, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Naghmeh Bahrami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Craniomaxillofacial Research Center, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Adnan Khosravi
- Tobacco Prevention and Control Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Kazem Pour Dizaji
- Biostatistics Department, Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Pathology Research Center, Dr Daneshbod Lab, Shiraz, Iran.,Shefa Neuroscience Research Center, Tehran, Iran
| |
Collapse
|
10
|
The Role of Hydrogen Peroxide and Peroxiredoxins throughout the Cell Cycle. Antioxidants (Basel) 2020; 9:antiox9040280. [PMID: 32224940 PMCID: PMC7222192 DOI: 10.3390/antiox9040280] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/22/2023] Open
Abstract
Hydrogen peroxide (H2O2) is an oxidizing agent that induces cellular damage at inappropriate concentrations and gives rise to an arrest during cell cycle progression, causing cell death. Recent evidence indicates that H2O2 also acts as a promoter for cell cycle progression by oxidizing specific thiol proteins. The intracellular concentration of H2O2 is regulated tightly, enabling its use as a cellular signaling molecule while minimizing its potential to cause cellular damage. Peroxiredoxins (Prxs) have peroxidase activity toward H2O2, organic hydroperoxides, and peroxynitrite for protecting cells from oxidative stress. They are suggested to work as signaling mediators, allowing the local accumulation of H2O2 by inactivating their peroxidase activity uniquely compared with other antioxidant proteins such as catalase and glutathione peroxidase. Given that Prxs are highly sensitive to oxidation by H2O2, they act as sensors and transducers of H2O2 signaling via transferring their oxidation state to effector proteins. The concentrations of intracellular H2O2 increase as the cell cycle progresses from G1 to mitosis. Here, we summarize the roles of Prxs with regard to the regulation of cell cycle-dependent kinase activity and anaphase-promoting complex/cyclosome in terms of changes in H2O2 levels. Protection of the cell from unwanted progression of the cell cycle is suggested to be a role of Prx. We discuss the possible roles of Prxs to control H2O2 levels.
Collapse
|
11
|
Byrne FL, Olzomer EM, Marriott GR, Quek LE, Katen A, Su J, Nelson ME, Hart-Smith G, Larance M, Sebesfi VF, Cuff J, Martyn GE, Childress E, Alexopoulos SJ, Poon IK, Faux MC, Burgess AW, Reid G, McCarroll JA, Santos WL, Quinlan KG, Turner N, Fazakerley DJ, Kumar N, Hoehn KL. Phenotypic screen for oxygen consumption rate identifies an anti-cancer naphthoquinone that induces mitochondrial oxidative stress. Redox Biol 2020; 28:101374. [PMID: 31743887 PMCID: PMC6861633 DOI: 10.1016/j.redox.2019.101374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 11/09/2022] Open
Abstract
A hallmark of cancer cells is their ability to reprogram nutrient metabolism. Thus, disruption to this phenotype is a potential avenue for anti-cancer therapy. Herein we used a phenotypic chemical library screening approach to identify molecules that disrupted nutrient metabolism (by increasing cellular oxygen consumption rate) and were toxic to cancer cells. From this screen we discovered a 1,4-Naphthoquinone (referred to as BH10) that is toxic to a broad range of cancer cell types. BH10 has improved cancer-selective toxicity compared to doxorubicin, 17-AAG, vitamin K3, and other known anti-cancer quinones. BH10 increases glucose oxidation via both mitochondrial and pentose phosphate pathways, decreases glycolysis, lowers GSH:GSSG and NAPDH/NAPD+ ratios exclusively in cancer cells, and induces necrosis. BH10 targets mitochondrial redox defence as evidenced by increased mitochondrial peroxiredoxin 3 oxidation and decreased mitochondrial aconitase activity, without changes in markers of cytosolic or nuclear damage. Over-expression of mitochondria-targeted catalase protects cells from BH10-mediated toxicity, while the thioredoxin reductase inhibitor auranofin synergistically enhances BH10-induced peroxiredoxin 3 oxidation and cytotoxicity. Overall, BH10 represents a 1,4-Naphthoquinone with an improved cancer-selective cytotoxicity profile via its mitochondrial specificity.
Collapse
Affiliation(s)
- Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| | - Ellen M Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Gabriella R Marriott
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Sydney, Australia
| | - Alice Katen
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| | - Jacky Su
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| | - Marin E Nelson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia
| | - Gene Hart-Smith
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Mark Larance
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia
| | - Veronica F Sebesfi
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jeff Cuff
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Gabriella E Martyn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Elizabeth Childress
- Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, VA, USA
| | - Stephanie J Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ivan K Poon
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Maree C Faux
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Antony W Burgess
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Glen Reid
- Concord Medical School, Asbestos Disease Research Institute, University of Sydney, Australia
| | - Joshua A McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Webster L Santos
- Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, VA, USA
| | - Kate Gr Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nigel Turner
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
12
|
Zhang S, He J, Tang M, Sun H. Prdx2 Upregulation Promotes the Growth and Survival of Gastric Cancer Cells. Pathol Oncol Res 2019; 26:1869-1877. [PMID: 31807984 DOI: 10.1007/s12253-019-00783-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022]
Abstract
Peroxiredoxins (Prdxs) play important roles in cell proliferation, differentiation, and the mediation of intracellular signalling pathways. Prdx2 is an important member of the peroxiredoxin family and is upregulated in many cancers. Until now, the biological functions of Prdx2 in gastric cancer have not been completely understood, and the underlying mechanisms remain elusive. The aim of this study was to identify the role of Prdx2 on the growth of gastric cancer cells and the underlying mechanisms. We demonstrated that Prdx2 was highly expressed in gastric cancer tissues and cell lines and that the over-expression of Prdx2 correlated with the progression of gastric cancer. Further, Prdx2 was silenced with a specific, lentiviral vector-mediated shRNA, and this suppressed the proliferation of gastric cancer cells and promoted the apoptosis of gastric cancer cells. Finally, the knockdown of Prdx2 contributed to the attenuated gastric cancer growth in BALB/c nude mice. In conclusion, these findings demonstrate that Prdx2 may participate in the carcinogenesis and development of gastric cancer.
Collapse
Affiliation(s)
- Shouru Zhang
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital , Chongqing, 400030, People's Republic of China
| | - Jingping He
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital , Chongqing, 400030, People's Republic of China
| | - Maocai Tang
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital , Chongqing, 400030, People's Republic of China
| | - Hao Sun
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital , Chongqing, 400030, People's Republic of China.
| |
Collapse
|
13
|
Cha HN, Park S, Dan Y, Kim JR, Park SY. Peroxiredoxin2 Deficiency Aggravates Aging-Induced Insulin Resistance and Declines Muscle Strength. J Gerontol A Biol Sci Med Sci 2019; 74:147-154. [PMID: 29733327 DOI: 10.1093/gerona/gly113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
This study examined the role of peroxiredoxin2 (Prx2) in aging-induced insulin resistance and reduction in skeletal muscle function in young (2-month-old) and old (24-month-old) Prx2 knockout (KO) and wild-type mice. Plasma insulin levels increased with aging in Prx2 KO mice but not in wild-type mice. Insulin sensitivity in the whole-body and skeletal muscle as assessed with the hyperinsulinemic-euglycemic clamp was lower in Prx2 KO mice than in wild-type mice in the old group but was not significantly different between the two genotypes in the young group. Insulin-induced activation of intracellular signaling molecules was also suppressed in old Prx2 KO mice compared to their wild-type littermates. Oxidative stress, inflammation, and p53 expression levels in skeletal muscle were higher in Prx2 KO mice than in wild-type mice in the old group but were not different between the two genotypes in the young group. p53 expression was negatively correlated with skeletal muscle insulin sensitivity in old mice. Skeletal muscle mass was similar between the two genotypes but grip strength was reduced in old Prx2 KO mice compared to old wild-type mice. These results suggest that Prx2 plays a protective role in aging-induced insulin resistance and declines in muscle strength by suppressing oxidative stress.
Collapse
Affiliation(s)
- Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Yongwook Dan
- Weinberg College of Art and Sciences, Northwestern University, Chicago, Illinois
| | - Jae-Ryong Kim
- Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Yeungnam University, Daegu, Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|
14
|
Sharapov MG, Novoselov VI. Catalytic and Signaling Role of Peroxiredoxins in Carcinogenesis. BIOCHEMISTRY (MOSCOW) 2019; 84:79-100. [PMID: 31216969 DOI: 10.1134/s0006297919020019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer cells experience strong oxidative stress caused by disorders in cell metabolism and action of external factors. For survival, cancer cells have developed a highly efficient system of antioxidant defense, some of the most important elements of which are peroxiredoxins (Prxs). Prxs are an evolutionarily ancient family of selenium-independent peroxidases that reduce a wide range of organic and inorganic hydroperoxides in the cell and the extracellular space. In addition, some Prxs exhibit chaperone and phospholipase activities. Prxs play an important role in the maintenance of the cell redox homeostasis; they prevent oxidation and aggregation of regulatory proteins, thereby affecting many cell signaling pathways. Prxs are involved in the regulation of cell growth, differentiation, and apoptosis. Due to their versatility and wide representation in all tissues and organs, Prxs participate in the development/suppression of many pathological conditions, among which cancer occupies a special place. This review focuses on the role of Prxs in the development of various forms of cancer. Understanding molecular mechanisms of Prx involvement in these processes will allow to develop new approaches to the prevention and treatment of cancer.
Collapse
Affiliation(s)
- M G Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - V I Novoselov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
15
|
Castaldo SA, Ajime T, Serrão G, Anastácio F, Rosa JT, Giacomantonio CA, Howarth A, Hill R, Madureira PA. Annexin A2 Regulates AKT Upon H₂O₂-Dependent Signaling Activation in Cancer Cells. Cancers (Basel) 2019; 11:cancers11040492. [PMID: 30959964 PMCID: PMC6520733 DOI: 10.3390/cancers11040492] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/31/2019] [Accepted: 04/03/2019] [Indexed: 11/16/2022] Open
Abstract
Hydrogen peroxide (H2O2) is a main second messenger in oncogenic signaling networks including the Ras and the growth factor receptor pathways. This is achieved predominantly through the oxidation of redox-sensitive cysteine (Cys) residues in proteins resulting in changes to their structure and function. We previously identified annexin A2 (ANXA2) as a redox regulatory protein that plays an important cellular role during oxidative stress and also promoting tumorigenesis. Here we investigated the role of ANXA2 in the regulation of H2O2-dependent signaling that drives tumor progression. We show that depletion of ANXA2 leads to the enhanced activation of AKT following either EGF/EGFR stimulation or oncogenic Ras transformation. The phosphatase and tensin homologue (PTEN) protein negatively regulates the PI3K/AKT pathway. We demonstrate that ANXA2 via its reactive Cys-8 residue, binds to PTEN and that the co-expression of PTEN and ANXA2, but not ANXA2 Cys-8-Ala mutant, inhibits AKT phosphorylation on Ser 473. These results indicate that ANXA2 is important for PTEN regulation within the PI3K/AKT signaling cascade. Furthermore, we also reveal that ANXA2 inversely regulates the expression of the peroxidase, peroxiredoxin 2, in a reactive oxygen species dependent manner.
Collapse
Affiliation(s)
- Stéphanie Anais Castaldo
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
| | - Tom Ajime
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
| | - Gisela Serrão
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
| | - Fábio Anastácio
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
| | - Joana Teixeira Rosa
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
| | | | - Alison Howarth
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, PO1 2DT Portsmouth, UK.
| | - Richard Hill
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, PO1 2DT Portsmouth, UK.
| | - Patrícia Alexandra Madureira
- Centre for Biomedical Research (CBMR), Campus of Gambelas, University of Algarve, Building 8, Room 2.22, 8005-139 Faro, Portugal.
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, PO1 2DT Portsmouth, UK.
| |
Collapse
|
16
|
Gu C, Luo J, Lu X, Tang Y, Ma Y, Yun Y, Cao J, Cao J, Huang Z, Zhou X, Zhang S. REV7 confers radioresistance of esophagus squamous cell carcinoma by recruiting PRDX2. Cancer Sci 2019; 110:962-972. [PMID: 30657231 PMCID: PMC6398896 DOI: 10.1111/cas.13946] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy has been widely used for the clinical management of esophageal squamous cell carcinoma. However, radioresistance remains a serious concern that prevents the efficacy of esophageal squamous cell carcinoma (ESCC) radiotherapy. REV7, the structural subunit of eukaryotic DNA polymerase ζ, has multiple functions in bypassing DNA damage and modulating mitotic arrest in human cell lines. However, the expression and molecular function of REV7 in ESCC progression remains unclear. In this study, we first examined the expression of REV7 in clinical ESCC samples, and we found higher expression of REV7 in ESCC tissues compared to matched adjacent or normal tissues. Knockdown of REV7 resulted in decreased colony formation and increased apoptosis in irradiated Eca‐109 and TE‐1 cells coupled with decreased tumor weight in a xenograft nude mouse model postirradiation. Conversely, overexpression of REV7 resulted in radioresistance in vitro and in vivo. Moreover, silencing of REV7 induced increased reactive oxygen species levels postirradiation. Proteomic analysis of REV7‐interacting proteins revealed that REV7 interacted with peroxiredoxin 2 (PRDX2), a well‐known antioxidant protein. Existence of REV7‐PRDX2 complex and its augmentation postirradiation were further validated by immunoprecipitation and immunofluorescence assays. REV7 knockdown significantly disrupted the presence of nuclear PRDX2 postirradiation, which resulted in oxidative stress. REV7‐PRDX2 complex also assembled onto DNA double‐strand breaks, whereas REV7 knockdown evidently increased double‐strand breaks that were unmerged by PRDX2. Taken together, the present study sheds light on REV7‐modulated radiosensitivity through interacting with PRDX2, which provides a novel target for ESCC radiotherapy.
Collapse
Affiliation(s)
- Cheng Gu
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Judong Luo
- Department of Oncology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xujing Lu
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Yiting Tang
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Yan Ma
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Yifei Yun
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Jianping Cao
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Juhua Cao
- Department of Internal Medicine, Changzhou No. 1 People's Hospital, Soochow University, Changzhou, China
| | - Zeyu Huang
- Department of Science and Education, Changzhou No. 3 People's Hospital, Changzhou, China
| | - Xifa Zhou
- Department of Radiation Oncology, Changzhou No. 4 People's Hospital, Soochow University, Changzhou, China
| | - Shuyu Zhang
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| |
Collapse
|
17
|
Cancer-Associated Function of 2-Cys Peroxiredoxin Subtypes as a Survival Gatekeeper. Antioxidants (Basel) 2018; 7:antiox7110161. [PMID: 30423872 PMCID: PMC6262534 DOI: 10.3390/antiox7110161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer cells are abnormal cells that do not comply with tissue homeostasis but undergo uncontrolled proliferation. Such abnormality is driven mostly by somatic mutations on oncogenes and tumor suppressors. Cancerous mutations show intra-tumoral heterogeneity across cancer types and eventually converge into the self-activation of proliferative signaling. While transient production of intracellular reactive oxygen species (ROS) is essential for cell signaling, its persistent production is cytotoxic. Thus, cancer cells require increased levels of intracellular ROS for continuous proliferation, but overexpress cellular peroxidase enzymes, such as 2-Cys peroxiredoxins, to maintain ROS homeostasis. However, suppression of 2-Cys peroxiredoxins has also been reported in some metastatic cancers. Hence, the cancer-associated functions of 2-Cys peroxiredoxins must be illuminated in the cellular context. In this review, we describe the distinctive signaling roles of 2-Cys peroxiredoxins beyond their intrinsic ROS-scavenging role in relation to cancer cell death and survival.
Collapse
|
18
|
Chandimali N, Jeong DK, Kwon T. Peroxiredoxin II Regulates Cancer Stem Cells and Stemness-Associated Properties of Cancers. Cancers (Basel) 2018; 10:cancers10090305. [PMID: 30177619 PMCID: PMC6162743 DOI: 10.3390/cancers10090305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/31/2018] [Accepted: 08/31/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells (CSCs) represent a sub-population of cancer cells with the ability to regulate stemness-associated properties which are specifically responsible for unlimited growth of cancers, generation of diverse cancer cells in differentiated state and resistance to existing chemotherapy and radiotherapy. Even though, current therapies destroy majority of cancer cells, it is believed to leave CSCs without eradicating which may be the conceptualization for chemoresistance and radio-resistance. Reactive oxygen species (ROS) maintain stem cells and regulate the stemness-associated properties of cancers. Beyond the maximum limit, ROS can damage cellular functions of cancers by subjecting them to oxidative stress. Thus, maintenance of ROS level plays an important role in cancers to regulate stemness-associated properties. Peroxiredoxin II (Prx II) is a member of peroxiredoxin antioxidant enzyme family which considers as a regulator of ROS in cellular environments by modulating redox status to maintain CSC phenotype and stemness properties. Prx II has cell type-dependent expression in various types of cancer cells and overexpression or silenced expression of Prx II in cancers is associated with stem cell phenotype and stemness-associated properties via activation or deactivation of various signaling pathways. In this review, we summarized available studies on Prx II expression in cancers and the mechanisms by which Prx II takes parts to regulate CSCs and stemness-associated properties. We further discussed the potential therapeutic effects of altering Prx II expression in cancers for better anticancer strategies by sensitizing cancer cells and stem cells to oxidative stress and inhibiting stemness-associated properties.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea.
| | - Dong Kee Jeong
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea.
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea.
| | - Taeho Kwon
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea.
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
19
|
Zhang Y, Lee JH, Paull TT, Gehrke S, D'Alessandro A, Dou Q, Gladyshev VN, Schroeder EA, Steyl SK, Christian BE, Shadel GS. Mitochondrial redox sensing by the kinase ATM maintains cellular antioxidant capacity. Sci Signal 2018; 11:eaaq0702. [PMID: 29991649 PMCID: PMC6042875 DOI: 10.1126/scisignal.aaq0702] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria are integral to cellular energy metabolism and ATP production and are involved in regulating many cellular processes. Mitochondria produce reactive oxygen species (ROS), which not only can damage cellular components but also participate in signal transduction. The kinase ATM, which is mutated in the neurodegenerative, autosomal recessive disease ataxia-telangiectasia (A-T), is a key player in the nuclear DNA damage response. However, ATM also performs a redox-sensing function mediated through formation of ROS-dependent disulfide-linked dimers. We found that mitochondria-derived hydrogen peroxide promoted ATM dimerization. In HeLa cells, ATM dimers were localized to the nucleus and inhibited by the redox regulatory protein thioredoxin 1 (TRX1), suggesting the existence of a ROS-mediated, stress-signaling relay from mitochondria to the nucleus. ATM dimer formation did not affect its association with chromatin in the absence or presence of nuclear DNA damage, consistent with the separation of its redox and DNA damage signaling functions. Comparative analysis of U2OS cells expressing either wild-type ATM or the redox sensing-deficient C2991L mutant revealed that one function of ATM redox sensing is to promote glucose flux through the pentose phosphate pathway (PPP) by increasing the abundance and activity of glucose-6-phosphate dehydrogenase (G6PD), thereby increasing cellular antioxidant capacity. The PPP produces the coenzyme NADPH needed for a robust antioxidant response, including the regeneration of TRX1, indicating the existence of a regulatory feedback loop involving ATM and TRX1. We propose that loss of the mitochondrial ROS-sensing function of ATM may cause cellular ROS accumulation and oxidative stress in A-T.
Collapse
Affiliation(s)
- Yichong Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ji-Hoon Lee
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Tanya T Paull
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Sarah Gehrke
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Qianhui Dou
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02155, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02155, USA
| | | | - Samantha K Steyl
- Department of Chemistry, Appalachian State University, Boone, NC 28608, USA
| | - Brooke E Christian
- Department of Chemistry, Appalachian State University, Boone, NC 28608, USA.
| | - Gerald S Shadel
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
20
|
Zhang Y, Wang D, Li M, Wei X, Liu S, Zhao M, Liu C, Wang X, Jiang X, Li X, Zhang S, Bergquist J, Wang B, Yang C, Mi J, Tian G. Quantitative Proteomics of TRAMP Mice Combined with Bioinformatics Analysis Reveals That PDGF-B Regulatory Network Plays a Key Role in Prostate Cancer Progression. J Proteome Res 2018; 17:2401-2411. [PMID: 29863873 DOI: 10.1021/acs.jproteome.8b00158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transgenic adenocarcinoma of the mouse prostate (TRAMP) mice is a widely used transgenic animal model of prostate cancer (PCa). We performed a label-free quantitative proteomics analysis combined with a bioinformatics analysis on the entire prostate protein extraction from TRAMP mice and compared it with WT littermates. From 2379 total identified proteins, we presented a modest mice prostate reference proteome containing 919 proteins. 61 proteins presented a significant expression difference between two groups. The integrative bioinformatics analysis predicted the overexpression of platelet-derived growth factor B (PDGF-B) in tumor tissues and supports the hypothesis of the PDGF-B signaling network as a key upstream regulator in PCa progression. Furthermore, we demonstrated that Crenolanib, a novel PDGF receptor inhibitor, inhibited PCa cell proliferation in a dose-dependent manner. Finally, we revealed the importance of PDGF-B regulatory network in PCa progression, which will assist us in understanding the role and mechanisms of PDGF-B in promoting cancer growth and provide valuable knowledge for future research on anti-PDGF therapy.
Collapse
Affiliation(s)
- Yuan Zhang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Dan Wang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Radiology , Affiliated Hospital of Binzhou Medical University , 661 Second Huanghe Road , Binzhou , Shandong Province 256603 , China
| | - Min Li
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Xiaodan Wei
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Shuang Liu
- College of Enology , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Miaoqing Zhao
- Department of Pathology , Provincial Hospital Affiliated to Shandong University , No. 324 Jingwu Weiqi Road , Jinan , Shandong Province 250021 , China
| | - Chu Liu
- Department of Urology , Yantai Yuhuangding Hospital , Zhifu District, No. 20, Yuhuangding East Road , Yantai , Shandong Province 264000 , China
| | - Xizhen Wang
- Imaging Center , Affiliated Hospital of Weifang Medical University , Kuiwen District, No. 465, Yuhe Road , Weifang , Shandong Province 256603 , China
| | - Xingyue Jiang
- Department of Radiology , Affiliated Hospital of Binzhou Medical University , 661 Second Huanghe Road , Binzhou , Shandong Province 256603 , China
| | - Xuri Li
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Shuping Zhang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Jonas Bergquist
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Chemistry - BMC , Uppsala University , P.O. Box 599, Husargatan 3 , Uppsala 75124 , Sweden
| | - Bin Wang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Chunhua Yang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Jia Mi
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Chemistry - BMC , Uppsala University , P.O. Box 599, Husargatan 3 , Uppsala 75124 , Sweden
| | - Geng Tian
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| |
Collapse
|
21
|
Angrisani A, Matrone N, Belli V, Vicidomini R, Di Maio N, Turano M, Scialò F, Netti PA, Porcellini A, Furia M. A functional connection between dyskerin and energy metabolism. Redox Biol 2017; 14:557-565. [PMID: 29132127 PMCID: PMC5684492 DOI: 10.1016/j.redox.2017.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023] Open
Abstract
The human DKC1 gene encodes dyskerin, an evolutionarily conserved nuclear protein whose overexpression represents a common trait of many types of aggressive sporadic cancers. As a crucial component of the nuclear H/ACA snoRNP complexes, dyskerin is involved in a variety of essential processes, including telomere maintenance, splicing efficiency, ribosome biogenesis, snoRNAs stabilization and stress response. Although multiple minor dyskerin splicing isoforms have been identified, their functions remain to be defined. Considering that low-abundance splice variants could contribute to the wide functional repertoire attributed to dyskerin, possibly having more specialized tasks or playing significant roles in changing cell status, we investigated in more detail the biological roles of a truncated dyskerin isoform that lacks the C-terminal nuclear localization signal and shows a prevalent cytoplasmic localization. Here we show that this dyskerin variant can boost energy metabolism and improve respiration, ultimately conferring a ROS adaptive response and a growth advantage to cells. These results reveal an unexpected involvement of DKC1 in energy metabolism, highlighting a previously underscored role in the regulation of metabolic cell homeostasis. Human dyskerin is an evolutionary conserved component of nuclear H/ACA snoRNPs. The functional role of a truncated dyskerin isoform (Iso3) is analyzed. Iso3 overexpression boosts energy metabolism and induces a ROS adaptive response. Iso3 connects dyskerin with mitochondrial functionality and redox homeostasis.
Collapse
Affiliation(s)
- Alberto Angrisani
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy.
| | - Nunzia Matrone
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy
| | - Valentina Belli
- Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci 53, 80125 Napoli, Italy
| | - Rosario Vicidomini
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy
| | - Nunzia Di Maio
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy
| | - Mimmo Turano
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy
| | - Filippo Scialò
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, University of Newcastle, Newcastle-upon-Tyne NE4 5PL, United Kingdom
| | - Paolo Antonio Netti
- Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci 53, 80125 Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, Università di Napoli Federico II, Piazzale Tecchio 80, 80125 Napoli, Italy
| | - Antonio Porcellini
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy
| | - Maria Furia
- Department of Biology, University of Naples "Federico II", Complesso Universitario Monte Santangelo, via Cinthia, 80126 Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, Università di Napoli Federico II, Piazzale Tecchio 80, 80125 Napoli, Italy.
| |
Collapse
|
22
|
Connor DE, Chaitanya GV, Chittiboina P, McCarthy P, Scott LK, Schrott L, Minagar A, Nanda A, Alexander JS. Variations in the cerebrospinal fluid proteome following traumatic brain injury and subarachnoid hemorrhage. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2017; 24:169-183. [PMID: 28549769 PMCID: PMC7303909 DOI: 10.1016/j.pathophys.2017.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Proteomic analysis of cerebrospinal fluid (CSF) has shown great promise in identifying potential markers of injury in neurodegenerative diseases [1-13]. Here we compared CSF proteomes in healthy individuals, with patients diagnosed with traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) in order to characterize molecular biomarkers which might identify these different clinical states and describe different molecular mechanisms active in each disease state. METHODS Patients presenting to the Neurosurgery service at the Louisiana State University Hospital-Shreveport with an admitting diagnosis of TBI or SAH were prospectively enrolled. Patients undergoing CSF sampling for diagnostic procedures were also enrolled as controls. CSF aliquots were subjected to 2-dimensional gel electrophoresis (2D GE) and spot percentage densities analyzed. Increased or decreased spot expression (compared to controls) was defined in terms of in spot percentages, with spots showing consistent expression change across TBI or SAH specimens being followed up by Matrix-Assisted Laser Desorption/Ionization mass spectrometry (MALDI-MS). Polypeptide masses generated were matched to known standards using a search of the NCBI and/or GenPept databases for protein matches. Eight hundred fifteen separately identifiable polypeptide migration spots were identified on 2D GE gels. MALDI-MS successfully identified 13 of 22 selected 2D GE spots as recognizable polypeptides. RESULTS Statistically significant changes were noted in the expression of fibrinogen, carbonic anhydrase-I (CA-I), peroxiredoxin-2 (Prx-2), both α and β chains of hemoglobin, serotransferrin (Tf) and N-terminal haptoglobin (Hp) in TBI and SAH specimens, as compared to controls. The greatest mean fold change among all specimens was seen in CA-I and Hp at 30.7 and -25.7, respectively. TBI specimens trended toward greater mean increases in CA-I and Prx-2 and greater mean decreases in Hp and Tf. CONCLUSIONS Consistent CSF elevation of CA-I and Prx-2 with concurrent depletion of Hp and Tf may represent a useful combination of biomarkers for the prediction of severity and prognosis following brain injury.
Collapse
Affiliation(s)
- David E Connor
- Baptist Health Neurosurgery Arkansas, Little Rock, AR, United States.
| | - Ganta V Chaitanya
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD, United States.
| | - Paul McCarthy
- Department of Medicine, Sect. of Nephrology, University of Maryland, Baltimore, MD, United States.
| | - L Keith Scott
- Department of Critical Care Medicine, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Lisa Schrott
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Anil Nanda
- Department of Neurosurgery, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| |
Collapse
|
23
|
Carlomosti F, D'Agostino M, Beji S, Torcinaro A, Rizzi R, Zaccagnini G, Maimone B, Di Stefano V, De Santa F, Cordisco S, Antonini A, Ciarapica R, Dellambra E, Martelli F, Avitabile D, Capogrossi MC, Magenta A. Oxidative Stress-Induced miR-200c Disrupts the Regulatory Loop Among SIRT1, FOXO1, and eNOS. Antioxid Redox Signal 2017; 27:328-344. [PMID: 27960536 DOI: 10.1089/ars.2016.6643] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS Reactive oxygen species (ROS) play a pivotal role in different pathologic conditions, including ischemia, diabetes, and aging. We previously showed that ROS enhance miR-200c expression, causing endothelial cell (EC) apoptosis and senescence. Herein, we dissect the interaction among miR-200c and three strictly related proteins that modulate EC function and ROS production: sirtuin 1 (SIRT1), endothelial nitric oxide synthase (eNOS), and forkhead box O1 (FOXO1). Moreover, the role of miR-200c on ROS modulation was also investigated. RESULTS We demonstrated that miR-200c directly targets SIRT1, eNOS, and FOXO1; via this mechanism, miR-200c decreased NO and increased the acetylation of SIRT1 targets, that is, FOXO1 and p53. FOXO1 acetylation inhibited its transcriptional activity on target genes, that is, SIRT1 and the ROS scavengers, catalase and manganese superoxide dismutase. In keeping, miR-200c increased ROS production and induced p66Shc protein phosphorylation in Ser-36; this mechanism upregulated ROS and inhibited FOXO1 transcription, reinforcing this molecular circuitry. These in vitro results were validated in three in vivo models of oxidative stress, that is, human skin fibroblasts from old donors, femoral arteries from old mice, and a murine model of hindlimb ischemia. In all cases, miR-200c was higher versus control and its targets, that is, SIRT1, eNOS, and FOXO1, were downmodulated. In the mouse hindlimb ischemia model, anti-miR-200c treatment rescued these targets and improved limb perfusion. Innovation and Conclusion: miR-200c disrupts SIRT1/FOXO1/eNOS regulatory loop. This event promotes ROS production and decreases NO, contributing to endothelial dysfunction under conditions of increased oxidative stress such as aging and ischemia. Antioxid. Redox Signal. 27, 328-344.
Collapse
Affiliation(s)
- Fabrizio Carlomosti
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Marco D'Agostino
- 2 Department of Experimental Medicine, University of Rome , Sapienza, Rome, Italy
| | - Sara Beji
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Alessio Torcinaro
- 3 Department of Biology and Biotechnology "Charles Darwin," Sapienza University , Rome, Italy .,4 Institute of Cell Biology and Neurobiology (IBCN) , National Research Council of Italy (CNR), Rome, Italy
| | - Roberto Rizzi
- 4 Institute of Cell Biology and Neurobiology (IBCN) , National Research Council of Italy (CNR), Rome, Italy
| | - Germana Zaccagnini
- 5 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato , Milan, Italy
| | - Biagina Maimone
- 5 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato , Milan, Italy
| | - Valeria Di Stefano
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Francesca De Santa
- 4 Institute of Cell Biology and Neurobiology (IBCN) , National Research Council of Italy (CNR), Rome, Italy .,6 Santa Lucia Foundation-FSL-IRCCS , Rome, Italy
| | - Sonia Cordisco
- 7 Molecular and Cell Biology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Annalisa Antonini
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Roberta Ciarapica
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Elena Dellambra
- 7 Molecular and Cell Biology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| | - Fabio Martelli
- 5 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato , Milan, Italy
| | - Daniele Avitabile
- 8 Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, IRCCS , Milan, Italy
| | | | - Alessandra Magenta
- 1 Vascular Pathology Laboratory, Istituto Dermopatico dell'Immacolata-IRCCS , FLMM, Rome, Italy
| |
Collapse
|
24
|
|
25
|
Benfeitas R, Uhlen M, Nielsen J, Mardinoglu A. New Challenges to Study Heterogeneity in Cancer Redox Metabolism. Front Cell Dev Biol 2017; 5:65. [PMID: 28744456 PMCID: PMC5504267 DOI: 10.3389/fcell.2017.00065] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are important pathophysiological molecules involved in vital cellular processes. They are extremely harmful at high concentrations because they promote the generation of radicals and the oxidation of lipids, proteins, and nucleic acids, which can result in apoptosis. An imbalance of ROS and a disturbance of redox homeostasis are now recognized as a hallmark of complex diseases. Considering that ROS levels are significantly increased in cancer cells due to mitochondrial dysfunction, ROS metabolism has been targeted for the development of efficient treatment strategies, and antioxidants are used as potential chemotherapeutic drugs. However, initial ROS-focused clinical trials in which antioxidants were supplemented to patients provided inconsistent results, i.e., improved treatment or increased malignancy. These different outcomes may result from the highly heterogeneous redox responses of tumors in different patients. Hence, population-based treatment strategies are unsuitable and patient-tailored therapeutic approaches are required for the effective treatment of patients. Moreover, due to the crosstalk between ROS, reducing equivalents [e.g., NAD(P)H] and central metabolism, which is heterogeneous in cancer, finding the best therapeutic target requires the consideration of system-wide approaches that are capable of capturing the complex alterations observed in all of the associated pathways. Systems biology and engineering approaches may be employed to overcome these challenges, together with tools developed in personalized medicine. However, ROS- and redox-based therapies have yet to be addressed by these methodologies in the context of disease treatment. Here, we review the role of ROS and their coupled redox partners in tumorigenesis. Specifically, we highlight some of the challenges in understanding the role of hydrogen peroxide (H2O2), one of the most important ROS in pathophysiology in the progression of cancer. We also discuss its interplay with antioxidant defenses, such as the coupled peroxiredoxin/thioredoxin and glutathione/glutathione peroxidase systems, and its reducing equivalent metabolism. Finally, we highlight the need for system-level and patient-tailored approaches to clarify the roles of these systems and identify therapeutic targets through the use of the tools developed in personalized medicine.
Collapse
Affiliation(s)
- Rui Benfeitas
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Jens Nielsen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| |
Collapse
|
26
|
Knockdown of PRDX2 sensitizes colon cancer cells to 5-FU by suppressing the PI3K/AKT signaling pathway. Biosci Rep 2017; 37:BSR20160447. [PMID: 28432271 PMCID: PMC5426286 DOI: 10.1042/bsr20160447] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/07/2017] [Accepted: 04/21/2017] [Indexed: 01/06/2023] Open
Abstract
Although, 5-Fluorouracil (5-FU) remains widely used in adjuvant therapy in patients with colon cancer, resistance to 5-FU-based chemotherapy is an important reason for treatment failure. Recent studies have reported that an enhanced reactive oxygen species (ROS) scavenging system shows drug resistance to 5-FU. Peroxiredoxin-2 (PRDX2), is an important member of the ROS scavenging system, and may be a potential target that promotes chemosensitivity to 5-FU in colon cancer. Here, we depleted PRDX2 by PRDX2-shRNA-LV transduction in two colon cancer cell lines and found that in vitro PRDX2 knockdown facilitates cell death, and apoptosis in 5-FU-treated colon cancer cells. In addition, we found that PRDX2 depletion in mice treated with 5-FU resulted in, inhibition of tumor growth, compared with mice treated with 5-FU alone. Our data also suggested that the PI3K/AKT signaling pathway links PRDX2 with 5-FU-induced apoptosis in colon cancer. Furthermore, when PRDX2 was overexpressed in colon cancer cells, we found increased p-AKT protein expression and reduced Bcl-2/Bax protein expression. PRDX2 and p-AKT protein expression were analyzed by immunohistochemistry technology in human colon carcinoma tissues. Pearson correlation coefficient is 0.873 and P<0.05. PRDX2 depletion led to reduced p-AKT expression and PI3K/AKT pathway inhibition promoted cell apoptosis in HT29 cell line. Taken together, our study suggests that decreasing the expression of PRDX2 could be a promising strategy for increasing the sensitivity of colon cancer cells to 5-FU.
Collapse
|
27
|
Yazdian-Robati R, Ahmadi H, Riahi MM, Lari P, Aledavood SA, Rashedinia M, Abnous K, Ramezani M. Comparative proteome analysis of human esophageal cancer and adjacent normal tissues. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:265-271. [PMID: 28392898 PMCID: PMC5378963 DOI: 10.22038/ijbms.2017.8354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective(s): Ranking as the sixth commonest cancer, esophageal squamous cell carcinoma (ESCC) represents one of the leading causes of cancer death worldwide. One of the main reasons for the low survival of patients with esophageal cancer is its late diagnosis. Materials and Methods: We used proteomics approach to analyze ESCC tissues with the aim of a better understanding of the malignant mechanism and searching candidate protein biomarkers for early diagnosis of esophageal cancer. The differential protein expression between cancerous and normal esophageal tissues was investigated by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE). Then proteins were identified by matrix-assisted laser desorption/ ionization tandem time-of-flight mass spectrometry (MALDI-TOF/TOF-MS) and MASCOT web based search engine. Results: We reported 4 differentially expressed proteins involved in the pathological process of esophageal cancer, such as annexinA1 (ANXA1), peroxiredoxin-2 (PRDX2), transgelin (TAGLN) andactin-aortic smooth muscle (ACTA2). Conclusion: In this report we have introduced new potential biomarker (ACTA2). Moreover, our data confirmed some already known markers for EC in our region.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Homa Ahmadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Matbou Riahi
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parisa Lari
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Amir Aledavood
- Cancer Research Center, Department of Radiation oncology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Rashedinia
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Nanotechnology Research Center, Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad Iran
| |
Collapse
|
28
|
Konno M, Asai A, Kawamoto K, Nishida N, Satoh T, Doki Y, Mori M, Ishii H. The one-carbon metabolism pathway highlights therapeutic targets for gastrointestinal cancer (Review). Int J Oncol 2017; 50:1057-1063. [PMID: 28259896 DOI: 10.3892/ijo.2017.3885] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 12/22/2016] [Indexed: 11/05/2022] Open
Abstract
After the initial use of anti-folates for treatment of malignancies, folate metabolism has emerged as a rational diagnostic and therapeutic target in gastrointestinal cancer. The one-carbon metabolic pathway, which comprises three critical reactions (i.e., folate and methionine cycles), underlies this effect in conjunction with the trans-sulfuration pathway. Understanding of the one-carbon metabolism pathway has served to unravel the link between the causes and effects of cancer phenotypes leading to several seminal discoveries such as that of diadenosine tri-phosphate hydrolase, microRNAs, 5-FU and, more recently, trifluridine. In the folate cycle, glycine and serine fuel the mitochondrial enzymes SHMT2, MTHFD2 and ALDH1L2, which play critical roles in the cancer survival and proliferation presumably through purine production. In the methionine cycle, S-adenocyl methionine serves hydrocarbons and polyamines that are critical for the epigenetic controls. The trans-sulfuration pathway is a critical component in the synthesis of glutathione, which is involved in the production of reactive oxygen species in cancer stem cells. Therefore, characterization of one-carbon metabolism is indispensable to the development of precision medicine in the context of cancer diagnostics and therapeutics. In the present study, we review the historical issues associated with one-carbon metabolism and highlight the recent advances in cancer research.
Collapse
Affiliation(s)
- Masamitsu Konno
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| | - Ayumu Asai
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| | - Koichi Kawamoto
- Department of Gastroenterological Surgery Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Naohiro Nishida
- Department of Gastroenterological Surgery Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Taroh Satoh
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
29
|
Park YH, Kim SU, Kwon TH, Kim JM, Song IS, Shin HJ, Lee BK, Bang DH, Lee SJ, Lee DS, Chang KT, Kim BY, Yu DY. Peroxiredoxin II promotes hepatic tumorigenesis through cooperation with Ras/Forkhead box M1 signaling pathway. Oncogene 2015; 35:3503-13. [PMID: 26500057 DOI: 10.1038/onc.2015.411] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/09/2015] [Accepted: 09/18/2015] [Indexed: 12/14/2022]
Abstract
The current study was carried out to define the involvement of Peroxiredoxin (Prx) II in progression of hepatocellular carcinoma (HCC) and the underlying molecular mechanism(s). Expression and function of Prx II in HCC was determined using H-ras(G12V)-transformed HCC cells (H-ras(G12V)-HCC cells) and the tumor livers from H-ras(G12V)-transgenic (Tg) mice and HCC patients. Prx II was upregulated in H-ras(G12V)-HCC cells and H-ras(G12V)-Tg mouse tumor livers, the expression pattern of which highly similar to that of forkhead Box M1 (FoxM1). Moreover, either knockdown of FoxM1 or site-directed mutagenesis of FoxM1-binding site of Prx II promoter significantly reduced Prx II levels in H-ras(G12V)-HCC cells, indicating FoxM1 as a direct transcription factor of Prx II in HCC. Interestingly, the null mutation of Prx II markedly decreased the number and size of tumors in H-ras(G12V)-Tg livers. Consistent with this, knockdown of Prx II in H-ras(G12V)-HCC cells reduced the expression of cyclin D1, cell proliferation, anchorage-independent growth and tumor formation in athymic nude mice, whereas overexpression of Prx II increased or aggravated the tumor phenotypes. Importantly, the expression of Prx II was correlated with that of FoxM1 in HCC patients. The activation of extracellular signal-related kinase (ERK) pathway and the expression of FoxM1 and cyclin D1 were highly dependent on Prx II in H-ras(G12V)-HCC cells and H-ras(G12V)-Tg livers. Prx II is FoxM1-dependently-expressed antioxidant in HCC and function as an enhancer of Ras(G12V) oncogenic potential in hepatic tumorigenesis through activation of ERK/FoxM1/cyclin D1 cascade.
Collapse
Affiliation(s)
- Y-H Park
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, Korea
| | - S-U Kim
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, Korea
| | - T-H Kwon
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - J-M Kim
- School of Medicine, Chungnam National University, Daejeon, Korea
| | - I-S Song
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - H-J Shin
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - B-K Lee
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - D-H Bang
- School of Medicine, Wonkwang University, Iksan, Korea
| | - S-J Lee
- Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - D-S Lee
- College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - K-T Chang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - B-Y Kim
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Korea
| | - D-Y Yu
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, Korea
| |
Collapse
|
30
|
Lim JM, Lee KS, Woo HA, Kang D, Rhee SG. Control of the pericentrosomal H2O2 level by peroxiredoxin I is critical for mitotic progression. J Cell Biol 2015; 210:23-33. [PMID: 26150388 PMCID: PMC4493999 DOI: 10.1083/jcb.201412068] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Reversible oxidative inactivation of centrosome-bound protein phosphatases such as Cdc14B by H2O2 is likely responsible for the inhibition of Cdk1-opposing phosphatases during early mitosis, which prevents premature degradation of mitotic activators. Proteins associated with the centrosome play key roles in mitotic progression in mammalian cells. The activity of Cdk1-opposing phosphatases at the centrosome must be inhibited during early mitosis to prevent premature dephosphorylation of Cdh1—an activator of the ubiquitin ligase anaphase-promoting complex/cyclosome—and the consequent premature degradation of mitotic activators. In this paper, we show that reversible oxidative inactivation of centrosome-bound protein phosphatases such as Cdc14B by H2O2 is likely responsible for this inhibition. The intracellular concentration of H2O2 increases as the cell cycle progresses. Whereas the centrosome is shielded from H2O2 through its association with the H2O2-eliminating enzyme peroxiredoxin I (PrxI) during interphase, the centrosome-associated PrxI is selectively inactivated through phosphorylation by Cdk1 during early mitosis, thereby exposing the centrosome to H2O2 and facilitating inactivation of centrosome-bound phosphatases. Dephosphorylation of PrxI by okadaic acid–sensitive phosphatases during late mitosis again shields the centrosome from H2O2 and thereby allows the reactivation of Cdk1-opposing phosphatases at the organelle.
Collapse
Affiliation(s)
- Jung Mi Lim
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Kyung S Lee
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD 20892
| | - Hyun Ae Woo
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Dongmin Kang
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Sue Goo Rhee
- Yonsei Biomedical Research Institute, Yonsei University, Seoul 120-749, South Korea
| |
Collapse
|
31
|
Redox Regulation in Cancer Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:750798. [PMID: 26273424 PMCID: PMC4529979 DOI: 10.1155/2015/750798] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/03/2015] [Accepted: 02/10/2015] [Indexed: 12/17/2022]
Abstract
Reactive oxygen species (ROS) and ROS-dependent (redox regulation) signaling pathways and transcriptional activities are thought to be critical in stem cell self-renewal and differentiation during growth and organogenesis. Aberrant ROS burst and dysregulation of those ROS-dependent cellular processes are strongly associated with human diseases including many cancers. ROS levels are elevated in cancer cells partially due to their higher metabolism rate. In the past 15 years, the concept of cancer stem cells (CSCs) has been gaining ground as the subpopulation of cancer cells with stem cell-like properties and characteristics have been identified in various cancers. CSCs possess low levels of ROS and are responsible for cancer recurrence after chemotherapy or radiotherapy. Unfortunately, how CSCs control ROS production and scavenging and how ROS-dependent signaling pathways contribute to CSCs function remain poorly understood. This review focuses on the role of redox balance, especially in ROS-dependent cellular processes in cancer stem cells (CSCs). We updated recent advances in our understanding of ROS generation and elimination in CSCs and their effects on CSC self-renewal and differentiation through modulating signaling pathways and transcriptional activities. The review concludes that targeting CSCs by manipulating ROS metabolism/dependent pathways may be an effective approach for improving cancer treatment.
Collapse
|
32
|
Kwon T, Rho JK, Lee JC, Park YH, Shin HJ, Cho S, Kang YK, Kim BY, Yoon DY, Yu DY. An important role for peroxiredoxin II in survival of A549 lung cancer cells resistant to gefitinib. Exp Mol Med 2015; 47:e165. [PMID: 26021759 PMCID: PMC4454996 DOI: 10.1038/emm.2015.24] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/22/2014] [Accepted: 01/19/2015] [Indexed: 12/27/2022] Open
Abstract
Redox adaptation is an important concept that explains the mechanisms by which cancer cells survive under persistent endogenous oxidative stress and become resistant to certain anticancer agents. To investigate this concept, we determined the expression levels of peroxiredoxins (Prxs), antioxidant enzymes in drug-resistant non-small cell lung carcinoma cells. Prx II was remarkably increased only in A549/GR (gefitinib-resistant) cells compared with A549 cells, consistent with methylation/demethylation. Prx II was highly methylated in the A549 cells but was demethylated in the A549/GR cells. The elevated expression of Prx II resulted in the downregulation of reactive oxygen species (ROS) and cell death and upregulation of cell cycle progression in the A549/GR cells. When Prx II mRNA in the A549/GR cells was knocked down, the levels of ROS and apoptosis were significantly recovered to the levels of the controls. In addition, signaling molecules involved in apoptosis were increased in the A549/GR-shPrx II cells. There was no difference in the expression of MAPK/ERK between the A549/GR cells and A549/GR-shPrx II cells, but the phosphorylation of JNK was increased in the A549/GR cells and was markedly decreased in the A549/GR-shPrx II cells. Colony number and tumor growth were significantly decreased in the A549/GR-shPrx II cells compared with the A549/GR cells. Our findings suggest that Prx II has an important role in cancer cell survival via the modulation of signaling molecules involved in apoptosis and the phosphorylation of JNK by the downregulation of ROS levels in A549/GR cells.
Collapse
Affiliation(s)
- Taeho Kwon
- 1] Disease Model Research Laboratory, Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea [2] Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Jin Kyung Rho
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Jae Cheol Lee
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Young-Ho Park
- Disease Model Research Laboratory, Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Hye-Jun Shin
- Disease Model Research Laboratory, Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Sunwha Cho
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Yong-Kook Kang
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Bo-Yeon Kim
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk, Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Dae-Yeul Yu
- Disease Model Research Laboratory, Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| |
Collapse
|
33
|
Varshney S, Bhadada SK, Arya AK, Sharma S, Behera A, Bhansali A, Rao SD. Changes in parathyroid proteome in patients with primary hyperparathyroidism due to sporadic parathyroid adenomas. Clin Endocrinol (Oxf) 2014; 81:614-20. [PMID: 24766412 DOI: 10.1111/cen.12479] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/09/2014] [Accepted: 04/20/2014] [Indexed: 11/28/2022]
Abstract
PURPOSE The pathogenesis of parathyroid tumours is only partially understood. A direct approach using proteomics could be a promising tool to increase our understanding of parathyroid tumorigenesis. The aim of the study was to investigate differentially expressed proteins to explore the underlying molecular basis of the disease and identify potential target proteins responsible for the genesis of adenoma. METHODS Proteins were extracted from adenomatous and normal parathyroid tissues. Differentially expressed proteins were separated by two-dimensional gel electrophoresis (2-D) and identified by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry. Statistical analysis was performed using spss 10.01 software. RESULTS Comparative analysis of the 2-D profiles of proteins isolated from adenomatous and normal parathyroid tissues showed 15 differentially expressed proteins, of which 11 were overexpressed. The characterized proteins were associated with diverse cellular functions including regulation of cell organization, programmed cell death, transcription and signal transduction. CONCLUSION The differentially expressed proteins in parathyroid adenomas may potentially serve as new targets to investigate the mechanisms of parathyroid adenoma transformation.
Collapse
Affiliation(s)
- Shweta Varshney
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | | | | | | | |
Collapse
|
34
|
Avitabile D, Ranieri D, Nicolussi A, D’Inzeo S, Capriotti AL, Genovese L, Proietti S, Cucina A, Coppa A, Samperi R, Bizzarri M, Laganà A, Torrisi MR. Peroxiredoxin 2 nuclear levels are regulated by circadian clock synchronization in human keratinocytes. Int J Biochem Cell Biol 2014; 53:24-34. [DOI: 10.1016/j.biocel.2014.04.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 04/08/2014] [Accepted: 04/29/2014] [Indexed: 12/16/2022]
|
35
|
Jeon YJ, Kim J, Lee DS, Shim JH, Seo KS, Chae JI. Phosphorylation of PrxII promotes JNK-dependent apoptosis in adult cloned pig kidney. Int J Biochem Cell Biol 2014; 53:352-60. [DOI: 10.1016/j.biocel.2014.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 12/19/2022]
|
36
|
FENG JIHONG, FU ZHONGXUE, GUO JINBAO, LU WEIDONG, WEN KUNMING, CHEN WANGSHENG, WANG HAO, WEI JILAI, ZHANG SHOURU. Overexpression of peroxiredoxin 2 inhibits TGF-β1-induced epithelial-mesenchymal transition and cell migration in colorectal cancer. Mol Med Rep 2014; 10:867-73. [DOI: 10.3892/mmr.2014.2316] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/28/2014] [Indexed: 11/06/2022] Open
|
37
|
2-cys peroxiredoxins: emerging hubs determining redox dependency of Mammalian signaling networks. Int J Cell Biol 2014; 2014:715867. [PMID: 24672551 PMCID: PMC3932224 DOI: 10.1155/2014/715867] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/25/2013] [Indexed: 01/28/2023] Open
Abstract
Mammalian cells have a well-defined set of antioxidant enzymes, which includes superoxide dismutases, catalase, glutathione peroxidases, and peroxiredoxins. Peroxiredoxins are the most recently identified family of antioxidant enzymes that catalyze the reduction reaction of peroxides, such as H2O2. In particular, typical 2-Cys peroxiredoxins are the featured peroxidase enzymes that receive the electrons from NADPH by coupling with thioredoxin and thioredoxin reductase. These enzymes distribute throughout the cellular compartments and, therefore, are thought to be broad-range antioxidant defenders. However, recent evidence demonstrates that typical 2-Cys peroxiredoxins play key signal regulatory roles in the various signaling networks by interacting with or residing near a specific redox-sensitive molecule. These discoveries help reveal the redox signaling landscape in mammalian cells and may further provide a new paradigm of therapeutic approaches based on redox signaling.
Collapse
|
38
|
Lu W, Fu Z, Wang H, Feng J, Wei J, Guo J. Peroxiredoxin 2 is upregulated in colorectal cancer and contributes to colorectal cancer cells' survival by protecting cells from oxidative stress. Mol Cell Biochem 2013; 387:261-70. [PMID: 24234423 DOI: 10.1007/s11010-013-1891-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/05/2013] [Indexed: 02/02/2023]
Abstract
Peroxiredoxin 2 (Prdx2) is a member of the peroxiredoxin family, which is responsible for neutralizing reactive oxygen species. Prdx2 has been found to be elevated in several human cancer cells and tissues, including colorectal cancer (CRC), and it influences diverse cellular processes involving cells' survival, proliferation, and apoptosis, which suggests a possible role for Prdx2 in the maintenance of cancer cell. However, the mechanism by which Prdx2 modulates CRC cells' survival is unknown. The current study aimed to determine the effect of elevated Prdx2 on CRC cells and to further understand the underlying mechanisms. The results of this study showed that Prdx2 was upregulated in CRC tissues compared with the matched noncancer colorectal mucosa tissues and that Prdx2 expression was positively associated with tumor metastasis and the TNM stage. In the LoVo CRC cell line, Prdx2 was upregulated at both the RNA and protein levels compared with the normal FHC colorectal mucosa cell line. In addition, the LoVo CRC cell line was significantly more resistant to hydrogen peroxide (H₂O₂)-induced apoptosis because of a failure to activate pro-apoptotic pathways in contrast to Prdx2 knockdown cells. Suppression of Prdx2 using a lentiviral vector-mediated Prdx2-specific shRNA in the LoVo cell line restored H₂O₂ sensitivity. Our results suggested that Prdx2 has an essential role in regulating oxidation-induced apoptosis in CRC cells. Prdx2 may have potential as a therapeutic target in CRC.
Collapse
|
39
|
Lu W, Fu Z, Wang H, Feng J, Wei J, Guo J. Peroxiredoxin 2 knockdown by RNA interference inhibits the growth of colorectal cancer cells by downregulating Wnt/β-catenin signaling. Cancer Lett 2013; 343:190-9. [PMID: 24125860 DOI: 10.1016/j.canlet.2013.10.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 11/24/2022]
Abstract
Peroxiredoxin 2 (Prdx2) has been shown to act as an antioxidant whose main function is to reduce hydrogen peroxide (H2O2) in cells, and Prdx2 is abnormally elevated in colorectal cancer (CRC). However, the functional significance of this up-regulation and the detailed molecular mechanism behind the regulatory effect of Prdx2 on the growth of CRC cells have not been elucidated. In this study, we demonstrated that Prdx2 knockdown using a lentiviral vector-mediated specific shRNA inhibited cell growth, stimulated apoptosis, and augmented the production of endogenous reactive oxygen species (ROS). Further, silencing of Prdx2 resulted in an altered expression of proteins associated with the Wnt signaling pathway. Finally, Prdx2 knockdown contributed to attenuated CRC growth in BALB/c nude mice. In conclusion, these findings demonstrate that the regulatory effects of Prdx2 can be partially attributed to Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Weidong Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhongxue Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| | - Hao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jihong Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jinlai Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jinbao Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
40
|
Haynes AC, Qian J, Reisz JA, Furdui CM, Lowther WT. Molecular basis for the resistance of human mitochondrial 2-Cys peroxiredoxin 3 to hyperoxidation. J Biol Chem 2013; 288:29714-23. [PMID: 24003226 DOI: 10.1074/jbc.m113.473470] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peroxiredoxins (Prxs) detoxify peroxides and modulate H2O2-mediated cell signaling in normal and numerous pathophysiological contexts. The typical 2-Cys subclass of Prxs (human Prx1-4) utilizes a Cys sulfenic acid (Cys-SOH) intermediate and disulfide bond formation across two subunits during catalysis. During oxidative stress, however, the Cys-SOH moiety can react with H2O2 to form Cys sulfinic acid (Cys-SO2H), resulting in inactivation. The propensity to hyperoxidize varies greatly among human Prxs. Mitochondrial Prx3 is the most resistant to inactivation, but the molecular basis for this property is unknown. A panel of chimeras and Cys variants of Prx2 and Prx3 were treated with H2O2 and analyzed by rapid chemical quench and time-resolved electrospray ionization-TOF mass spectrometry. The latter utilized an on-line rapid-mixing setup to collect data on the low seconds time scale. These approaches enabled the first direct observation of the Cys-SOH intermediate and a putative Cys sulfenamide (Cys-SN) for Prx2 and Prx3 during catalysis. The substitution of C-terminal residues in Prx3, residues adjacent to the resolving Cys residue, resulted in a Prx2-like protein with increased sensitivity to hyperoxidation and decreased ability to form the intermolecular disulfide bond between subunits. The corresponding Prx2 chimera became more resistant to hyperoxidation. Taken together, the results of this study support that the kinetics of the Cys-SOH intermediate is key to determine the probability of hyperoxidation or disulfide formation. Given the oxidizing environment of the mitochondrion, it makes sense that Prx3 would favor disulfide bond formation as a protection mechanism against hyperoxidation and inactivation.
Collapse
Affiliation(s)
- Alexina C Haynes
- From the Center for Structural Biology and Department of Biochemistry
| | | | | | | | | |
Collapse
|
41
|
Jiang W, Bian L, Wang N, He Y. Proteomic analysis of protein expression profiles during hyperthermia-induced apoptosis in Tca8113 cells. Oncol Lett 2013; 6:135-143. [PMID: 23946791 PMCID: PMC3742465 DOI: 10.3892/ol.2013.1354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 04/18/2013] [Indexed: 11/15/2022] Open
Abstract
The aim of the present study was to explore protein expression profiles during cancer cell apoptosis induced by hyperthermia. A hyperthermia-induced apoptosis model was established using a Tca8113 cell line derived from a human tongue squamous cell carcinoma, which underwent fluorescent differential display two-dimensional (2D) gel electrophoresis at 2, 6, 8, 12 and 24 h following the induction of hyperthermia. Proteins were identified by mass spectrometry analysis. Expression changes in the proteins were detected by western blot analysis. A total of 107 proteins were detected that exhibited different expression levels in the hyperthermia-treated cells compared with the controls, and 57 of these proteins were identified. Expression changes in the representative proteins were further verified by western blot analysis. These 57 proteins were identified according to the following functional groups: energy metabolism-related enzymes, cytoskeleton-related proteins, chaperones, transcription factors, protein synthesis-related proteins and cell division- and proliferation-related proteins. These groups included 44 upregulated and 13 downregulated proteins. Among the 44 upregulated proteins, 27 were upregulated continuously, eight were upregulated at an early time-point and nine were upregulated at a middle to late time-point. Among the 13 downregulated proteins, five were downregulated continuously, six were downregulated at an early time-point and two were downregulated at a middle to late time-point. These results indicate that hyperthermia-induced Tca8113 cell apoptosis is controlled by multiple factors, which include time and regulatory proteins.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan 650031; ; The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000
| | | | | | | |
Collapse
|
42
|
Peroxiredoxin-3 is overexpressed in prostate cancer and promotes cancer cell survival by protecting cells from oxidative stress. Br J Cancer 2013; 109:983-93. [PMID: 23880827 PMCID: PMC3749568 DOI: 10.1038/bjc.2013.396] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 06/19/2013] [Accepted: 06/24/2013] [Indexed: 12/31/2022] Open
Abstract
Objective: We have previously identified peroxiredoxin-3 (PRDX-3) as a cell-surface protein that is androgen regulated in the LNCaP prostate cancer (PCa) cell line. PRDX-3 is a member of the peroxiredoxin family that are responsible for neutralising reactive oxygen species. Experimental design: PRDX-3 expression was examined in tissue from 32 patients using immunohistochemistry. Subcellular distribution was determined using confocal microscopy. PRDX-3 expression was determined in antiandrogen-resistant cell lines by western blotting and quantitative RT–PCR. The pathways of PRDX-3 overexpression and knockdown on apoptosis and response to oxidative stress were investigated using protein arrays. Results: PRDX-3 is upregulated in a number of endocrine-regulated tumours; in particular in PCa and prostatic intraepithelial neoplasia. Although the majority of PRDX-3 is localised to the mitochondria, we have confirmed that PRDX-3 at the cell membrane is androgen regulated. In antiandrogen-resistant LNCaP cell lines, PRDX-3 is upregulated at the protein but not RNA level. Resistant cells also possess an upregulation of the tricarboxylic acid (TCA) pathway and resistance to H2O2-induced apoptosis through a failure to activate pro-apoptotic pathways. Knockdown of PRDX-3 restored H2O2 sensitivity. Conclusion: Our results suggest that PRDX-3 has an essential role in regulating oxidation-induced apoptosis in antiandrogen-resistant cells. PRDX-3 may have potential as a therapeutic target in castrate-independent PCa.
Collapse
|
43
|
Lee DJ, Kang DH, Choi M, Choi YJ, Lee JY, Park JH, Park YJ, Lee KW, Kang SW. Peroxiredoxin-2 represses melanoma metastasis by increasing E-Cadherin/β-Catenin complexes in adherens junctions. Cancer Res 2013; 73:4744-57. [PMID: 23749642 DOI: 10.1158/0008-5472.can-12-4226] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In melanoma, transition to the vertical growth phase is the critical step in conversion to a deadly malignant disease. Here, we offer the first evidence that an antioxidant enzyme has a key role in this transition. We found that the antioxidant enzyme peroxiredoxin-2 (Prx2) inversely correlated with the metastatic capacity of human melanoma cells. Silencing Prx2 expression stimulated proliferation and migration, whereas ectopic expression of Prx2 produced the opposite effect. Mechanistic investigations indicated that Prx2 negatively regulated Src/ERK activation status, which in turn fortified adherens junctions function by increasing E-cadherin expression and phospho-Y654-dependent retention of β-catenin in the plasma membrane. In murine melanoma cells, Prx2 silencing enhanced lung metastasis in vivo. Interestingly, the natural compound gliotoxin, which is known to exert a Prx-like activity, inhibited proliferation and migration as well as lung metastasis of Prx2-deficient melanoma cells. Overall, our findings reveal that Prx2 is a key regulator of invasion and metastasis in melanoma, and also suggest a pharmacologic strategy to effectively decrease deadly malignant forms of this disease.
Collapse
Affiliation(s)
- Doo Jae Lee
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Borger E, Aitken L, Du H, Zhang W, Gunn-Moore FJ, Du Yan SS. Is amyloid binding alcohol dehydrogenase a drug target for treating Alzheimer's disease? Curr Alzheimer Res 2013; 10:21-29. [PMID: 22742981 PMCID: PMC3674883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 06/12/2012] [Accepted: 06/27/2012] [Indexed: 06/01/2023]
Abstract
Current strategies for the treatment of Alzheimer's disease (AD) involve tackling the formation or clearance of the amyloid-beta peptide (Aβ) and/or hyper-phosphorylated tau, or the support and stabilization of the remaining neuronal networks. However, as we gain a clearer idea of the large number of molecular mechanisms at work in this disease, it is becoming clearer that the treatment of AD should take a combined approach of dealing with several aspects of the pathology. The concept that we also need to protect specific sensitive targets within the cell should also be considered. In particular the role of protecting the function of a specific mitochondrial protein, amyloid binding alcohol dehydrogenase (ABAD), will be the focus of this review. Mitochondrial dysfunction is a well-recognized fact in the progression of AD, though until recently the mechanisms involved could only be loosely labeled as changes in 'metabolism'. The discovery that Aβ can be present within the mitochondria and specifically bind to ABAD, has opened up a new area of AD research. Here we review the evidence that the prevention of Aβ binding to ABAD is a drug target for the treatment of AD.
Collapse
Affiliation(s)
- Eva Borger
- School of Biology, Medical and Biological Sciences Building, North Haugh, University of St Andrews, Scotland UK KY16 9TF
| | - Laura Aitken
- School of Biology, Medical and Biological Sciences Building, North Haugh, University of St Andrews, Scotland UK KY16 9TF
| | - Heng Du
- Higuchi Bioscience Center and Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS66047, USA
| | - Wenshen Zhang
- State Key Laboratory of Earth Surface Process and Resource Ecology, Center for Natural Medicine Engineering, The Ministry of Education of China, Beijing Normal University, Beijing 100875, China
| | - Frank J Gunn-Moore
- School of Biology, Medical and Biological Sciences Building, North Haugh, University of St Andrews, Scotland UK KY16 9TF
| | - Shirley Shi Du Yan
- Higuchi Bioscience Center and Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS66047, USA
| |
Collapse
|
45
|
Yan S, Chen S, Li Z, Wang H, Huang T, Wang X, Wang J. Biochemical characterization of human peroxiredoxin 2, an antioxidative protein. Acta Biochim Biophys Sin (Shanghai) 2012; 44:759-64. [PMID: 22805285 DOI: 10.1093/abbs/gms061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human peroxiredoxin 2 (Prx2), which is abundant in erythrocytes, has been shown to play a key role in protecting erythrocytes against oxidative stress by scavenging reactive oxygen species as well as participating in cell signal transduction. Here, human Prx2 gene was successfully cloned into Escherichia coli BL21 (DE3) for Prx2 expression. Sodium dodecyl sulfate polyacrylamide gel electrophoresis analysis suggested that the recombinant protein was expressed mainly in a soluble form. The recombinant protein was purified by one-step Ni-nitrilotriacetic acid chelating affinity chromatography to a purity of up to 91.5%. The peroxidase activity of Prx2 to scavenge H(2)O(2) was determined by a ferrithiocyanate assay. The ability of Prx2 to protect plasmid DNA was tested by using a mixed-function oxidation system, and results showed that Prx2 could prevent DNA from undergoing oxidative stress. Ultraviolet (UV)-induced cell apoptosis assay demonstrated that Prx2 is also able to protect NIH/3T3 cells from UV-induced damage, suggesting its possible applications in cosmetics and other areas.
Collapse
Affiliation(s)
- Sheng Yan
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Reactive oxygen species mediated DNA damage is essential for abnormal erythropoiesis in peroxiredoxin II−/− mice. Biochem Biophys Res Commun 2012; 424:189-95. [DOI: 10.1016/j.bbrc.2012.06.113] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 06/21/2012] [Indexed: 01/02/2023]
|
47
|
Peroxiredoxin 2 specifically regulates the oxidative and metabolic stress response of human metastatic breast cancer cells in lungs. Oncogene 2012; 32:724-35. [PMID: 22430214 DOI: 10.1038/onc.2012.93] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Little is known about metastatic pathways that are specific to the lung rather than other organs. We previously showed that antioxidant proteins such as peroxiredoxins were specifically upregulated in lung metastatic breast cancer cells. We hypothesize that cancer cells that live under aerobic conditions, as might be the case in lungs, protect themselves against the damage caused by reactive oxygen species (ROS). To examine this hypothesis, we studied the role of peroxiredoxin-2 (PRDX2) in lung vs bone metastasis formation. A metastatic variant of MDA-MB-435 breast cancer cells that specifically metastasize to lungs (435-L3) was transduced with short hairpin RNAs to specifically silence PRDX2. Conversely, a bone metastatic variant of MDA-MB-231 cells (BO2) was stably transfected to overexpress PRDX2. The 435-L3 cells silenced for PRDX2 were significantly more sensitive to H(2)O(2)-induced oxidative stress than the parental and scrambled transfected cells. BO2/PRDX2 cells produced less ROS than BO2/green fluorescent protein control cells under oxidative stress. Moreover, PRDX2 knockdown inhibited the growth of 435-L3 cells in the lungs, whereas lymph node metastasis remained unaffected. In contrast, PRDX2 overexpression in bone metastatic BO2 breast cancer cells led to drastic inhibition of the skeletal tumor burden and reduction of bone destruction. Furthermore, PRDX2 expression in breast cancer cells was associated with a glucose-dependent phenotype, different from bone metastatic cells. Overall, our results strongly suggest that PRDX2 is a targetable 'metabolic adaptor' driver protein implicated in the selective growth of metastatic cells in the lungs by protecting them against oxidative stress.
Collapse
|
48
|
Chae HZ, Oubrahim H, Park JW, Rhee SG, Chock PB. Protein glutathionylation in the regulation of peroxiredoxins: a family of thiol-specific peroxidases that function as antioxidants, molecular chaperones, and signal modulators. Antioxid Redox Signal 2012; 16:506-23. [PMID: 22114845 PMCID: PMC3270059 DOI: 10.1089/ars.2011.4260] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
SIGNIFICANCE Reversible protein glutathionylation plays an important role in cellular regulation, signaling transduction, and antioxidant defense. This redox-sensitive mechanism is involved in regulating the functions of peroxiredoxins (Prxs), a family of ubiquitously expressed thiol-specific peroxidase enzymes. Glutathionylation of certain Prxs at their active-site cysteines not only provides reducing equivalents to support their peroxidase activity but also protects Prxs from irreversible hyperoxidation. Typical 2-Cys Prx also functions as a molecular chaperone when it exists as a decamer and/or higher molecular weight complexes. The hyperoxidized sulfinic derivative of 2-Cys Prx is reactivated by sulfiredoxin (Srx). In this review, the roles of glutathionylation in the regulation of Prxs are discussed with respect to their molecular structure and functions as antioxidants, molecular chaperones, and signal modulators. RECENT ADVANCES Recent findings reveal that glutathionylation regulates the quaternary structure of Prx. Glutathionylation of Prx I at Cys(83) converts the decameric Prx to its dimers with the loss of chaperone activity. The findings that dimer/oligomer structure specific Prx I binding proteins, e.g., phosphatase and tensin homolog (PTEN) and mammalian Ste20-like kinase-1 (MST1), regulate cell cycle and apoptosis, respectively, suggest a possible link between glutathionylation and those signaling pathways. CRITICAL ISSUES Knowing how glutathionylation affects the interaction between Prx I and its nearly 20 known interacting proteins, e.g., PTEN and MST1 kinase, would reveal new insights on the physiological functions of Prx. FUTURE DIRECTIONS In vitro studies reveal that Prx oligomerization is linked to its functional changes. However, in vivo dynamics, including the effect by glutathionylation, and its physiological significance remain to be investigated.
Collapse
Affiliation(s)
- Ho Zoon Chae
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | | | | | | | | |
Collapse
|
49
|
Ishii T, Warabi E, Yanagawa T. Novel roles of peroxiredoxins in inflammation, cancer and innate immunity. J Clin Biochem Nutr 2012; 50:91-105. [PMID: 22448089 PMCID: PMC3303482 DOI: 10.3164/jcbn.11-109] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 09/20/2011] [Indexed: 02/06/2023] Open
Abstract
Peroxiredoxins possess thioredoxin or glutathione peroxidase and chaperone-like activities and thereby protect cells from oxidative insults. Recent studies, however, reveal additional functions of peroxiredoxins in gene expression and inflammation-related biological reactions such as tissue repair, parasite infection and tumor progression. Notably, peroxiredoxin 1, the major mammalian peroxiredoxin family protein, directly interacts with transcription factors such as c-Myc and NF-κB in the nucleus. Additionally, peroxiredoxin 1 is secreted from some cells following stimulation with TGF-β and other cytokines and is thus present in plasma and body fluids. Peroxiredoxin 1 is now recognized as one of the pro-inflammatory factors interacting with toll-like receptor 4, which triggers NF-κB activation and other signaling pathways to evoke inflammatory reactions. Some cancer cells release peroxiredoxin 1 to stimulate toll-like receptor 4-mediated signaling for their progression. Interestingly, peroxiredoxins expressed in protozoa and helminth may modulate host immune responses partly through toll-like receptor 4 for their survival and progression in host. Extracellular peroxiredoxin 1 and peroxiredoxin 2 are known to enhance natural killer cell activity and suppress virus-replication in cells. Peroxiredoxin 1-deficient mice show reduced antioxidant activities but also exhibit restrained tissue inflammatory reactions under some patho-physiological conditions. Novel functions of peroxiredoxins in inflammation, cancer and innate immunity are the focus of this review.
Collapse
Affiliation(s)
- Tetsuro Ishii
- Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | | |
Collapse
|