101
|
Walter L, Canup B, Pujada A, Bui TA, Arbasi B, Laroui H, Merlin D, Garg P. Matrix metalloproteinase 9 (MMP9) limits reactive oxygen species (ROS) accumulation and DNA damage in colitis-associated cancer. Cell Death Dis 2020; 11:767. [PMID: 32943603 PMCID: PMC7498454 DOI: 10.1038/s41419-020-02959-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Colitis-associated cancer (CAC) is a subtype of colon cancer that is driven by chronic inflammation and is prevalent in chronic ulcerative colitis patients. The development of CAC is associated with the inflammation-dysplasia-carcinoma pathway which is significantly different than adenoma-carcinoma pathway of sporadic colon cancer (CRC). Matrix Metalloproteinase 9 (MMP9) is a zinc-dependent endopeptidase against extracellular matrix (ECM) proteins expressed in the gastrointestinal tract during inflammation. We have previously shown that MMP9 plays a tumor suppressor role in CAC via “MMP9-Notch1-ARF-p53 axis” pathway. The aim of this study is to determine the role of MMP9 in maintaining genomic stability in CAC. Homozygous transgenic mice with constitutive-expression of MMP9 in the colonic epithelium (TgM9) with their wild-type littermates (WT) and stably transfected HCT116 cells with/without MMP9 were used for in vivo and in vitro experiments, respectively. As ‘proof of concept’ model, nanoparticles (NPs) loaded with MMP9 siRNA were used to examine the effect of MMP9 silencing in the colonic epithelium. In CAC, colonic epithelium of TgM9 mice exhibited lower amounts of reactive oxygen species (ROS), less DNA damage, and increased expression of mismatch repair genes compared to WTs. Our study showed that MMP9 expression correlates with the reduced ROS levels, decreased DNA damage, and upregulated mismatch repair pathway. This suggests that MMP9 expression is a natural biological way to suppress CAC by limiting ROS accumulation and DNA damage in the colon. Therefore, MMP9 inhibition could be deleterious for CAC patient.
Collapse
Affiliation(s)
- Lewins Walter
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Brandon Canup
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Adani Pujada
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Tien Anh Bui
- Department of Biology, Georgia State University, Atlanta, GA, United States
| | - Behafarin Arbasi
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Hamed Laroui
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Didier Merlin
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Pallavi Garg
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States.
| |
Collapse
|
102
|
Kuznietsova H, Dziubenko N, Herheliuk T, Prylutskyy Y, Tauscher E, Ritter U, Scharff P. Water-Soluble Pristine C 60 Fullerene Inhibits Liver Alterations Associated with Hepatocellular Carcinoma in Rat. Pharmaceutics 2020; 12:pharmaceutics12090794. [PMID: 32842595 PMCID: PMC7559840 DOI: 10.3390/pharmaceutics12090794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive production of reactive oxygen species is the main cause of hepatocellular carcinoma (HCC) initiation and progression. Water-soluble pristine C60 fullerene is a powerful and non-toxic antioxidant, therefore, its effect under rat HCC model and its possible mechanisms were aimed to be discovered. Studies on HepG2 cells (human HCC) demonstrated C60 fullerene ability to inhibit cell growth (IC50 = 108.2 μmol), to induce apoptosis, to downregulate glucose-6-phosphate dehydrogenase, to upregulate vimentin and p53 expression and to alter HepG2 redox state. If applied to animals experienced HCC in dose of 0.25 mg/kg per day starting at liver cirrhosis stage, C60 fullerene improved post-treatment survival similar to reference 5-fluorouracil (31 and 30 compared to 17 weeks) and inhibited metastasis unlike the latter. Furthermore, C60 fullerene substantially attenuated liver injury and fibrosis, decreased liver enzymes, and normalized bilirubin and redox markers (elevated by 1.7–7.7 times under HCC). Thus, C60 fullerene ability to inhibit HepG2 cell growth and HCC development and metastasis and to improve animal survival was concluded. C60 fullerene cytostatic action might be realized through apoptosis induction and glucose-6-phosphate dehydrogenase downregulation in addition to its antioxidant activity.
Collapse
Affiliation(s)
- Halyna Kuznietsova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
- Correspondence: (H.K.); (U.R.); Tel.: +38-095-277-4370 (H.K.); +49-3677-69-3603 (U.R.)
| | - Natalia Dziubenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Tetiana Herheliuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Yuriy Prylutskyy
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Eric Tauscher
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
- Correspondence: (H.K.); (U.R.); Tel.: +38-095-277-4370 (H.K.); +49-3677-69-3603 (U.R.)
| | - Peter Scharff
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
| |
Collapse
|
103
|
Chen P, Xiao Z, Wu H, Wang Y, Su W, Li P. The Effects of Naringin on Cigarette Smoke-Induced Dynamic Changes in Oxidation/Antioxidant System in Lung of Mice. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20947233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Naringin possesses strong antioxidative activity and can protect against some respiratory diseases. Oxidative stress is thought to be a major factor in the development of many tobacco-caused diseases. The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway plays a critical role in the regulation of oxidative stress. The dynamic changes in the antioxidant system in the lung that are induced by cigarette smoke (CS) are not well investigated, and how naringin affects these changes remains unknown. This study aimed to investigate the dynamic changes between the oxidation and antioxidant systems resulting from CS exposure and the effects of naringin on these changes in mice. Mice were chronically exposed to CS for 30 days. The levels of malondialdehyde (MDA), glutathione (GSH), interleukin (IL)-6, and tumor necrosis factor-alpha (TNF-α); the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px); and the expressions of Nrf2, heme oxygenase-1 (HO-1), and nicotinamide adenine dinucleotide phosphate quinone dehydrogenase 1 (NQO1) in lung tissue were measured on days 2, 7, and 30. The levels of MDA, GSH, IL-6, and TNF-α in the lung were found to increase throughout the exposure. SOD and GSH-Px activities showed an increase on day 2 and a decrease on days 7 and 30. The messenger ribonucleic acid expressions of Nrf2, HO-1, and NQO1 were elevated on day 2 and decreased on day 7; Nrf2 and HO-1 expressions were continually decreased, but NQO1 expression was increased again, on day 30. Naringin restored the levels of these biochemical indices to normal throughout the experiment, suggesting that naringin protected against the CS-induced oxidative damage by suppressing the increase of antioxidants resulting from the early stage of CS exposure, as well as inhibiting the depletion of antioxidants due to long-term oxidative stress. Naringin also suppressed lung inflammation by inhibiting IL-6 and TNF-α. These results indicate that naringin possesses a powerful ability to maintain the balance of the oxidation/antioxidant system in the lung when subjected to CS exposure, probably by regulating the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Pan Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ziting Xiao
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yonggang Wang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, Department of Ecology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
104
|
ROS-Mediated Therapeutic Strategy in Chemo-/Radiotherapy of Head and Neck Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5047987. [PMID: 32774675 PMCID: PMC7396055 DOI: 10.1155/2020/5047987] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/26/2020] [Indexed: 12/24/2022]
Abstract
Head and neck cancer is a highly genetic and metabolic heterogeneous collection of malignancies of the lip, oral cavity, salivary glands, pharynx, esophagus, paranasal sinuses, and larynx with five-year survival rates ranging from 12% to 93%. Patients with head and neck cancer typically present with advanced stage III, IVa, or IVb disease and are treated with comprehensive modality including chemotherapy, radiotherapy, and surgery. Despite advancements in treatment modality and technique, noisome recurrence, invasiveness, and resistance as well as posttreatment complications severely influence survival rate and quality of life. Thus, new therapeutic strategies are urgently needed that offer enhanced efficacy with less toxicity. ROS in cancer cells plays a vital role in regulating cell death, DNA repair, stemness maintenance, metabolic reprogramming, and tumor microenvironment, all of which have been implicated in resistance to chemo-/radiotherapy of head and neck cancer. Adjusting ROS generation and elimination to reverse the resistance of cancer cells without impairing normal cells show great hope in improving the therapeutic efficacy of chemo-/radiotherapy of head and neck cancer. In the current review, we discuss the pivotal and targetable redox-regulating system including superoxide dismutases (SODs), tripeptide glutathione (GSH), thioredoxin (Trxs), peroxiredoxins (PRXs), nuclear factor erythroid 2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/keap1), and mitochondria electron transporter chain (ETC) complexes and their roles in regulating ROS levels and their clinical significance implicated in chemo-/radiotherapy of head and neck cancer. We also summarize several old drugs (referred to as the non-anti-cancer drugs used in other diseases for a long time) and small molecular compounds as well as natural herbs which effectively modulate cellular ROS of head and neck cancer to synergize the efficacy of conventional chemo-/radiotherapy. Emerging interdisciplinary techniques including photodynamic, nanoparticle system, and Bio-Electro-Magnetic-Energy-Regulation (BEMER) therapy are promising measures to broaden the potency of ROS modulation for the benefit of chemo-/radiotherapy in head and neck cancer.
Collapse
|
105
|
Molecular characterization, purification, and antioxidant activity of recombinant superoxide dismutase from the Pacific abalone Haliotis discus hannai Ino. World J Microbiol Biotechnol 2020; 36:115. [PMID: 32661581 PMCID: PMC7359182 DOI: 10.1007/s11274-020-02892-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 07/07/2020] [Indexed: 12/19/2022]
Abstract
Superoxide dismutase (SOD) is an acidic metalloenzyme that scavenges free radicals produced by endogenous and exogenous substances. In the present study, the tissue distribution of the superoxide dismutase HdhCu/Zn-SOD was investigated in Haliotis discus hannai Ino. The expression profile after lipopolysaccharide (LPS) challenge was determined using quantitative real-time polymerase chain reaction (qPCR). To study the antioxidant activity of a recombinant HdhCu/Zn-SOD protein, the HdhCu/Zn-SOD gene was cloned into the pPIC9K vector and transformed into the Pichia pastoris GS115 strain by electroporation. After induction by methanol, the recombinant product was purified using immobilized metal affinity chromatography and confirmed using mass spectrometry. The optimal expression conditions were determined to be incubation with 0.5% methanol at pH 6.0, resulting in a stable expressed product with the molecular weight of approximately 17 kDa and 21 kDa. The enzymatic activity of HdhCu/Zn-SOD consistently increased with increasing Cu2+ concentrations and showed good thermal stability. Recombinant HdhCu/Zn-SOD showed a strong ability to scavenge superoxide anions and hydroxyl radicals and protected L929 cells against the toxicity caused by H2O2 through its in vitro antioxidant activity. The heterologous expression of HdhCu/Zn-SOD in P. pastoris and the antioxidant activity of this enzyme are reported for the first time.
Collapse
|
106
|
Narayanan D, Ma S, Özcelik D. Targeting the Redox Landscape in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12071706. [PMID: 32605023 PMCID: PMC7407119 DOI: 10.3390/cancers12071706] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are produced predominantly by the mitochondrial electron transport chain and by NADPH oxidases in peroxisomes and in the endoplasmic reticulum. The antioxidative defense counters overproduction of ROS with detoxifying enzymes and molecular scavengers, for instance, superoxide dismutase and glutathione, in order to restore redox homeostasis. Mutations in the redox landscape can induce carcinogenesis, whereas increased ROS production can perpetuate cancer development. Moreover, cancer cells can increase production of antioxidants, leading to resistance against chemo- or radiotherapy. Research has been developing pharmaceuticals to target the redox landscape in cancer. For instance, inhibition of key players in the redox landscape aims to modulate ROS production in order to prevent tumor development or to sensitize cancer cells in radiotherapy. Besides the redox landscape of a single cell, alternative strategies take aim at the multi-cellular level. Extracellular vesicles, such as exosomes, are crucial for the development of the hypoxic tumor microenvironment, and hence are explored as target and as drug delivery systems in cancer therapy. This review summarizes the current pharmaceutical and experimental interventions of the cancer redox landscape.
Collapse
Affiliation(s)
- Dilip Narayanan
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Sana Ma
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Dennis Özcelik
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
- current address: Chemistry | Biology | Pharmacy Information Center, ETH Zürich, Vladimir-Prelog-Weg 10, 8093 Zürich, Switzerland
- Correspondence:
| |
Collapse
|
107
|
Relationship between the antiproliferative properties of Cu(II) complexes with the Schiff base derived from pyridine-2-carboxaldehyde and 5,6-diamino-1,3-dimethyluracil and the redox status mediated by antioxidant defense systems on glioma tumoral cells. J Inorg Biochem 2020; 207:111053. [DOI: 10.1016/j.jinorgbio.2020.111053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 02/06/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022]
|
108
|
Serum levels of superoxide dismutases in patients with benign paroxysmal positional vertigo. Biosci Rep 2020; 40:224098. [PMID: 32391546 PMCID: PMC7240196 DOI: 10.1042/bsr20193917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 11/17/2022] Open
Abstract
Objective: To investigate the possible role of superoxide dismutases (SODs) in the development of benign paroxysmal positional vertigo (BPPV) and recurrence events in a 1-year follow-up study. Methods: This was a prospective one-center study. A total of 204 patients with BPPV and 120 age-and sex matched healthy subjects were included. The levels of SOD between patients and control cases were compared. The levels of SOD between posterior semicircular canal (PSC) and horizontal semicircular canal (HSC) were also compared. In the 1-year follow-up, recurrence events were confirmed. The influence of SOD levels on BPPV and recurrent BPPV were performed by binary logistic regression analysis. Results: The serum levels of SOD in patients with BPPV were lower than in those control cases (P<0.001). Levels of SOD did not differ in patients with PSC and HSC (P=0.42). As a categorical variable, for per interquartile range (IQR) increment of serum level of SOD, the unadjusted and adjusted risks of BPPV would be decreased by 72% (with the odds ratio [OR] of 0.28 [95% confidence interval (CI): 0.21–0.37], P<0.001) and 43% (0.57 [0.42–0.69], P<0.001), respectively. Recurrent attacks of BPPV were reported in 50 patients (24.5%). Patients with recurrent BPPV had lower levels of SOD than in patients without (P<0.001). For per IQR increment of serum level of SOD, the unadjusted and adjusted risks of BPPV would be decreased by 51% (with the OR of 0.49 [95% CI: 0.36–0.68], P<0.001) and 24% (0.76 [0.60–0.83], P<0.001), respectively. Conclusion: Reduced serum levels of SOD were associated with higher risk of BPPV and BPPV recurrence events.
Collapse
|
109
|
Shi C, Li Y, Gu N. Iron-Based Nanozymes in Disease Diagnosis and Treatment. Chembiochem 2020; 21:2722-2732. [PMID: 32315111 DOI: 10.1002/cbic.202000094] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/21/2020] [Indexed: 12/15/2022]
Abstract
Iron-based nanozymes are currently one of the few clinical inorganic nanoparticles for disease diagnosis and treatment. Overcoming the shortcomings of natural enzymes, such as easy inactivation and low yield, combined with their special nanometer properties and magnetic functions, iron-based nanozymes have broad prospects in biomedicine. This minireview summarizes their preparation, biological activity, catalytic mechanism, and applications in diagnosis and treatment of diseases. Finally, challenges to their future development and the trends of iron-based nanozymes are discussed. The purpose of this minireview is to better understand and reasonably speculate on the rational design of iron-based nanozymes as an increasingly important new paradigm for diagnostics.
Collapse
Affiliation(s)
- Chu Shi
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Yan Li
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
110
|
Stephenie S, Chang YP, Gnanasekaran A, Esa NM, Gnanaraj C. An insight on superoxide dismutase (SOD) from plants for mammalian health enhancement. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103917] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
111
|
Moldogazieva NT, Mokhosoev IM, Mel'nikova TI, Zavadskiy SP, Kuz'menko AN, Terentiev AA. Dual Character of Reactive Oxygen, Nitrogen, and Halogen Species: Endogenous Sources, Interconversions and Neutralization. BIOCHEMISTRY (MOSCOW) 2020; 85:S56-S78. [PMID: 32087054 DOI: 10.1134/s0006297920140047] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxidative stress resulting from accumulation of reactive oxygen, nitrogen, and halogen species (ROS, RNS, and RHS, respectively) causes the damage of cells and biomolecules. However, over the long evolutionary time, living organisms have developed the mechanisms for adaptation to oxidative stress conditions including the activity of the antioxidant system (AOS), which maintains low intracellular levels of RONS (ROS and RNS) and RHS. Moreover, living organisms have adapted to use low concentrations of these electrophiles for the regulation of cell functions through the reversible post-translational chemical modifications of redox-sensitive amino acid residues in intracellular effectors of signal transduction pathways (protein kinases and protein phosphatases), transcription factors, etc. An important fine-tuning mechanism that ensures involvement of RONS and RHS in the regulation of physiological processes is interconversion between different reactive species. This review focuses on the complex networks of interacting RONS and RHS types and their endogenous sources, such as NOX family of NADPH oxidases, complexes I and III of the mitochondrial electron transport chain, NO synthases, cytochrome P450-containing monooxygenase system, xanthine oxidoreductase, and myeloperoxidases. We highlight that kinetic parameters of reactions involving RONS and RHS determine the effects of these reactive species on cell functions. We also describe the functioning of enzymatic and non-enzymatic AOS components and the mechanisms of RONS and RHS scavenging under physiological conditions. We believe that analysis of interactions between RONS and relationships between different endogenous sources of these compounds will contribute to better understanding of their role in the maintenance of cell redox homeostasis as well as initiation and progression of diseases.
Collapse
Affiliation(s)
- N T Moldogazieva
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| | - I M Mokhosoev
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia.
| | - T I Mel'nikova
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - S P Zavadskiy
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - A N Kuz'menko
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - A A Terentiev
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
112
|
Beneficial Regulation of Cellular Oxidative Stress Effects, and Expression of Inflammatory, Angiogenic, and the Extracellular Matrix Remodeling Proteins by 1α,25-Dihydroxyvitamin D3 in a Melanoma Cell Line. Molecules 2020; 25:molecules25051164. [PMID: 32150881 PMCID: PMC7179240 DOI: 10.3390/molecules25051164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 12/29/2022] Open
Abstract
The causes of cancer include the cellular accumulation reactive oxygen species (ROS), which overrides the cellular antioxidants such as superoxide dismutase, from intrinsic aging, genetics, and exposure to environmental pollutants and ultraviolet (UV) radiation. The ROS damage biomolecules such as DNA (including p53 gene), RNA, and lipids, and activate inflammatory, angiogenic, and extracellular matrix (ECM) remodeling proteins; which collectively facilitate carcinogenesis. The 1α,25-dihydroxyvitamin D3 (Vitamin D) has anti-carcinogenic potential from its antioxidant, anti-inflammatory, and endocrine properties. We examined the anti-carcinogenic mechanism of vitamin D through the beneficial regulation of oxidative stress effects (oxidative DNA/RNA damage, superoxide dismutase expression, membrane damage, and p53 promoter activity), and expression (at the protein, mRNA and/or promoter levels) of inflammatory mediators (interleukin-1 (IL-1) and tumor necrosis factor-α (TNF-α)), angiogenic mediators (transforming growth factor-β (TGF-β), and vascular endothelial growth factor (VEGF)), and the ECM remodeling proteins (matrix metalloproteinases (MMP)-1 and MMP-2) by vitamin D in melanoma cells. Vitamin D inhibited oxidative DNA/RNA damage and membrane damage; and stimulated superoxide dismutase expression and p53 promoter activity in melanoma cells. It inhibited the expression of IL-1, TNF-α, TGF-β, VEGF, MMP-1 and MMP-2 by transcriptional or post-transcriptional mechanisms. We conclude that vitamin D is beneficial to melanoma cells through the inhibition of oxidative DNA/RNA damage, membrane damage, and the expression of inflammatory, angiogenic and ECM remodeling proteins; and the stimulation of superoxide dismutase expression and p53 promoter activity.
Collapse
|
113
|
Jahangir Chughtai MF, Pasha I, Zahoor T, Khaliq A, Ahsan S, Wu Z, Nadeem M, Mehmood T, Amir RM, Yasmin I, Liaqat A, Tanweer S. Nutritional and therapeutic perspectives of Stevia rebaudiana as emerging sweetener; a way forward for sweetener industry. CYTA - JOURNAL OF FOOD 2020. [DOI: 10.1080/19476337.2020.1721562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Imran Pasha
- National Institute of Food Science & Technology, Faculty of Food Nutrition & Home Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Tahir Zahoor
- National Institute of Food Science & Technology, Faculty of Food Nutrition & Home Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Adnan Khaliq
- Department of Food Science & Technology, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Pakistan
| | - Samreen Ahsan
- Department of Food Science & Technology, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Pakistan
| | - Zhengzhong Wu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS University Islamabad, Vehari, Punjab, Pakistan
| | - Tariq Mehmood
- Department of Food Science & Technology, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Pakistan
| | - Rai Muhammad Amir
- Institute of Food and Nutritional Sciences, PMAS Arid Agriculture University, Rawalpindi, Punjab, Pakistan
| | - Iqra Yasmin
- Department of Food Science and Technology, Government College Women University, Faisalabad, Pakistan
- Barani Agricultural Research Institute, Chakwal, Pakistan
| | - Atif Liaqat
- Department of Food Science & Technology, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Pakistan
| | - Saira Tanweer
- Department of Food Science and Technology, University College of Agricultural and Environmental Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
114
|
Santoro V, Kovalenko I, Vriens K, Christen S, Bernthaler A, Haegebarth A, Fendt SM, Christian S. SLC25A32 sustains cancer cell proliferation by regulating flavin adenine nucleotide (FAD) metabolism. Oncotarget 2020; 11:801-812. [PMID: 32166001 PMCID: PMC7055544 DOI: 10.18632/oncotarget.27486] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/16/2019] [Indexed: 12/30/2022] Open
Abstract
SLC25A32 is a member of the solute carrier 25 family of mitochondrial transporters. SLC25A32 transports tetrahydrofolate (THF) as well as FAD into mitochondria and regulates mitochondrial one-carbon metabolism and redox balance. While it is known that cancer cells require one-carbon and FAD-dependent mitochondrial metabolism to sustain cell proliferation, the role of SLC25A32 in cancer cell growth remains unexplored. Our results indicate that the SLC25A32 gene is highly amplified in different tumors and that amplification correlates with increased mRNA expression and reduced patients´ survival. siRNA-mediated knock-down and CRISPR-mediated knock-out of SLC25A32 in cancer cells of different origins, resulted in the identification of cell lines sensitive and resistant to SLC25A32 inhibition. Mechanistically, tracing of deuterated serine revealed that SLC25A32 knock-down does not affect the mitochondrial/cytosolic folate flux as measured by Liquid Chromatography coupled Mass Spectrometry (LC-MS). Instead, SLC25A32 inhibition results in a respiratory chain dysfunction at the FAD-dependent complex II enzyme, induction of Reactive Oxygen Species (ROS) and depletion of reduced glutathione (GSH), which impairs cancer cell proliferation. Moreover, buthionine sulfoximine (BSO) treatment further sensitizes cells to ROS-mediated inhibition of cell proliferation upon SLC25A32 knock-down. Treatment of cells with the FAD precursor riboflavin and with GSH rescues cancer cell proliferation upon SLC25A32 down-regulation. Our results indicate that the reduction of mitochondrial FAD concentrations by targeting SLC25A32 has potential clinical applications as a single agent or in combination with approved cancer drugs that lead to increased oxidative stress and reduced tumor growth.
Collapse
Affiliation(s)
| | - Ilya Kovalenko
- Bayer AG, Drug Discovery, Berlin 13353, Germany.,Current address: University of Michigan, Cancer Center, Ann Arbor, MI 48108, USA
| | - Kim Vriens
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium.,Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven 3000, Belgium
| | - Stefan Christen
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium.,Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven 3000, Belgium
| | | | | | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium.,Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven 3000, Belgium
| | | |
Collapse
|
115
|
Sisakht M, Darabian M, Mahmoodzadeh A, Bazi A, Shafiee SM, Mokarram P, Khoshdel Z. The role of radiation induced oxidative stress as a regulator of radio-adaptive responses. Int J Radiat Biol 2020; 96:561-576. [PMID: 31976798 DOI: 10.1080/09553002.2020.1721597] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose: Various sources of radiation including radiofrequency, electromagnetic radiation (EMR), low- dose X-radiation, low-level microwave radiation and ionizing radiation (IR) are indispensable parts of modern life. In the current review, we discussed the adaptive responses of biological systems to radiation with a focus on the impacts of radiation-induced oxidative stress (RIOS) and its molecular downstream signaling pathways.Materials and methods: A comprehensive search was conducted in Web of Sciences, PubMed, Scopus, Google Scholar, Embase, and Cochrane Library. Keywords included Mesh terms of "radiation," "electromagnetic radiation," "adaptive immunity," "oxidative stress," and "immune checkpoints." Manuscripts published up until December 2019 were included.Results: RIOS induces various molecular adaptors connected with adaptive responses in radiation exposed cells. One of these adaptors includes p53 which promotes various cellular signaling pathways. RIOS also activates the intrinsic apoptotic pathway by depolarization of the mitochondrial membrane potential and activating the caspase apoptotic cascade. RIOS is also involved in radiation-induced proliferative responses through interaction with mitogen-activated protein kinases (MAPks) including p38 MAPK, ERK, and c-Jun N-terminal kinase (JNK). Protein kinase B (Akt)/phosphoinositide 3-kinase (PI3K) signaling pathway has also been reported to be involved in RIOS-induced proliferative responses. Furthermore, RIOS promotes genetic instability by introducing DNA structural and epigenetic alterations, as well as attenuating DNA repair mechanisms. Inflammatory transcription factors including macrophage migration inhibitory factor (MIF), nuclear factor κB (NF-κB), and signal transducer and activator of transcription-3 (STAT-3) paly major role in RIOS-induced inflammation.Conclusion: In conclusion, RIOS considerably contributes to radiation induced adaptive responses. Other possible molecular adaptors modulating RIOS-induced responses are yet to be divulged in future studies.
Collapse
Affiliation(s)
- Mohsen Sisakht
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Darabian
- Department of Radiology, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Bazi
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Sayed Mohammad Shafiee
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khoshdel
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
116
|
Plasma proteomics-based identification of novel biomarkers in early gastric cancer. Clin Biochem 2020; 76:5-10. [PMID: 31765635 DOI: 10.1016/j.clinbiochem.2019.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/27/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022]
|
117
|
Seelig J, Heller RA, Hackler J, Haubruck P, Moghaddam A, Biglari B, Schomburg L. Selenium and copper status - potential signposts for neurological remission after traumatic spinal cord injury. J Trace Elem Med Biol 2020; 57:126415. [PMID: 31685353 DOI: 10.1016/j.jtemb.2019.126415] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/23/2019] [Accepted: 10/05/2019] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Traumatic Spinal Cord Injury (TSCI) is a severe incident resulting in loss of motor and sensory function caused by complex pathological mechanisms including massive oxidative stress and extensive inflammatory processes. The essential trace elements selenium (Se) and copper (Cu) play crucial roles as part of the antioxidant defense. HYPOTHESIS Remission after TSCI is associated with characteristic dynamics of early changes in serum Cu and Se status. STUDY DESIGN Single-center prospective observational study. PATIENTS AND METHODS Serum samples from TSCI patients were analyzed (n = 52); 21 recovered and showed a positive abbreviated injury score (AIS) conversion within 3 months (G1), whereas 21 had no remission (G0). Ten subjects with vertebral fractures without neurological impairment served as control (C). Different time points (at admission, and after 4, 9, 12, and 24 h) were analyzed for total serum Se and Cu concentrations by total reflection X-ray fluorescence, and for Selenoprotein P (SELENOP) and Ceruloplasmin (CP) by sandwich ELISA. RESULTS At admission, CP and SELENOP concentrations were higher in the remission group (G1) than in the non-remission group (G0). Within 24 h, there were marginal changes in Se, SELENOP, Cu and CP concentrations in the groups of controls (C) and G0. In contrast, these parameters decreased significantly in G1. Binary logistic regression analysis including Cu and Se levels at admission in combination with Se and CP levels after 24 h allowed a prediction for potential remission, with an area under the curve (AUC) of 87.7% (CI: 75.1%-100.0%). CONCLUSION These data indicate a strong association between temporal changes of the Se and Cu status and the clinical outcome after TSCI. The dynamics observed may reflect an ongoing redistribution of the trace elements in favor of a better anti-inflammatory response and a more successful neurological regeneration.
Collapse
Affiliation(s)
- Julian Seelig
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Raban Arved Heller
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Heidelberg Trauma Research Group, Department of Trauma and Reconstructive Surgery, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Hackler
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Patrick Haubruck
- Heidelberg Trauma Research Group, Department of Trauma and Reconstructive Surgery, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Heidelberg, Germany; Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney, St Leonards, New South Wales, 2065, Australia
| | - Arash Moghaddam
- Aschaffenburg Trauma and Orthopedic Research Group, Center for Orthopedics, Trauma Surgery and Sports Medicine, Hospital Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | - Bahram Biglari
- BG Trauma Center Ludwigshafen, Department of Paraplegiology, Ludwigshafen, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
118
|
Lian S, Zhao L, Xun X, Lou J, Li M, Li X, Wang S, Zhang L, Hu X, Bao Z. Genome-Wide Identification and Characterization of SODs in Zhikong Scallop Reveals Gene Expansion and Regulation Divergence after Toxic Dinoflagellate Exposure. Mar Drugs 2019; 17:md17120700. [PMID: 31842317 PMCID: PMC6949909 DOI: 10.3390/md17120700] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
Abstract
As filter-feeding animals mainly ingesting microalgae, bivalves could accumulate paralytic shellfish toxins (PSTs) produced by harmful algae through diet. To protect themselves from the toxic effects of PSTs, especially the concomitant oxidative damage, the production of superoxide dismutase (SOD), which is the only eukaryotic metalloenzyme capable of detoxifying superoxide, may assist with toxin tolerance in bivalves. To better understand this process, in the present study, we performed the first systematic analysis of SOD genes in bivalve Chlamys farreri, an important aquaculture species in China. A total of six Cu/Zn-SODs (SOD1-6) and two Mn-SODs (SOD7, SOD8) were identified in C. farreri, with gene expansion being revealed in Cu/Zn-SODs. In scallops exposed to two different PSTs-producing dinoflagellates, Alexandrium minutum and A. catenella, expression regulation of SOD genes was analyzed in the top ranked toxin-rich organs, the hepatopancreas and the kidney. In hepatopancreas, which mainly accumulates the incoming PSTs, all of the six Cu/Zn-SODs showed significant alterations after A. minutum exposure, with SOD1, 2, 3, 5, and 6 being up-regulated, and SOD4 being down-regulated, while no significant change was detected in Mn-SODs. After A. catenella exposure, up-regulation was observed in SOD2, 4, 6, and 8, and SOD7 was down-regulated. In the kidney, where PSTs transformation occurs, SOD4, 5, 6, and 8 were up-regulated, and SOD7 was down-regulated in response to A. minutum feeding. After A. catenella exposure, all the Cu/Zn-SODs except SOD1 were up-regulated, and SOD7 was down-regulated in kidney. Overall, in scallops after ingesting different toxic algae, SOD up-regulation mainly occurred in the expanded Cu/Zn-SOD group, and SOD6 was the only member being up-regulated in both toxic organs, which also showed the highest fold change among all the SODs, implying the importance of SOD6 in protecting scallops from the stress of PSTs. Our results suggest the diverse function of scallop SODs in response to the PST-producing algae challenge, and the expansion of Cu/Zn-SODs might be implicated in the adaptive evolution of scallops or bivalves with respect to antioxidant defense against the ingested toxic algae.
Collapse
Affiliation(s)
- Shanshan Lian
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Liang Zhao
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Xiaogang Xun
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
| | - Jiarun Lou
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
| | - Moli Li
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
| | - Xu Li
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
| | - Shi Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Lingling Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Xiaoli Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: ; Tel.: +86-0532-8203-1970; Fax: +86-0532-8203-1802
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266003, China; (S.L.); (L.Z.); (X.X.); (J.L.); (M.L.); (X.L.); (S.W.); (L.Z.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
119
|
Mei S, Song X, Wang Y, Wang J, Su S, Zhu J, Geng Y. Studies on Protection of Astaxanthin from Oxidative Damage Induced by H 2O 2 in RAW 264.7 Cells Based on 1H NMR Metabolomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13568-13576. [PMID: 31709793 DOI: 10.1021/acs.jafc.9b04587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astaxanthin (AST) is a fat-soluble and non-vitamin A source of carotenoid that can quench reactive oxygen species and it has strong antioxidant and anti-inflammatory abilities. Herein, we have used H2O2 to establish a model of oxidative damage to RAW 264.7 cells and cells treated with vitamin C as the positive control group. The changes in metabolome were examined using 1H NMR and the results demonstrated that H2O2 treatment and various metabolic pathways such as amino acid, glucose, and glycerolipid metabolism were downregulated, which in turn affected citric acid cycle and energy status. AST could reverse downregulation of some of these metabolic pathways to a certain extent, and reduce cellular oxidative stress and death. The AST group differed from the vitamin C group in regulating d-glutamine, d-glutamic acid, pyruvate, and glycerolipid metabolism. The experimental results help to further understand the antioxidant effects of AST.
Collapse
Affiliation(s)
- Suhuan Mei
- Key Laboratory of Food Nutrition and Safety of SDNU, Provincial Key Laboratory of Animal Resistant Biology, College of Life Science , Shandong Normal University , Jinan 250014 , China
| | - Xiao Song
- Key Laboratory of Food Nutrition and Safety of SDNU, Provincial Key Laboratory of Animal Resistant Biology, College of Life Science , Shandong Normal University , Jinan 250014 , China
| | - Yali Wang
- Key Laboratory of Food Nutrition and Safety of SDNU, Provincial Key Laboratory of Animal Resistant Biology, College of Life Science , Shandong Normal University , Jinan 250014 , China
| | - Jun Wang
- Shandong Institute for Food and Drug Control , Jinan 250101 , China
| | - Shufang Su
- Shandong Institute for Food and Drug Control , Jinan 250101 , China
| | - Jianhua Zhu
- Shandong Institute for Food and Drug Control , Jinan 250101 , China
| | - Yue Geng
- Key Laboratory of Food Nutrition and Safety of SDNU, Provincial Key Laboratory of Animal Resistant Biology, College of Life Science , Shandong Normal University , Jinan 250014 , China
| |
Collapse
|
120
|
Plotnikova MA, Klotchenko SA, Kiselev AA, Gorshkov AN, Shurygina APS, Vasilyev KA, Uciechowska-Kaczmarzyk U, Samsonov SA, Kovalenko AL, Vasin AV. Meglumine acridone acetate, the ionic salt of CMA and N-methylglucamine, induces apoptosis in human PBMCs via the mitochondrial pathway. Sci Rep 2019; 9:18240. [PMID: 31796757 PMCID: PMC6890692 DOI: 10.1038/s41598-019-54208-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/04/2019] [Indexed: 01/16/2023] Open
Abstract
Meglumine acridone acetate (MA) is used in Russia for the treatment of influenza and other acute respiratory viral infections. It was assumed, until recently, that its antiviral effect was associated with its potential ability to induce type I interferon. Advanced studies, however, have shown the failure of 10-carboxymethyl-9-acridanone (CMA) to activate human STING. As such, MA's antiviral properties are still undergoing clarification. To gain insight into MA's mechanisms of action, we carried out RNA-sequencing analysis of global transcriptomes in MA-treated (MA+) human peripheral blood mononuclear cells (PBMCs). In response to treatment, approximately 1,223 genes were found to be differentially expressed, among which 464 and 759 were identified as either up- or down-regulated, respectively. To clarify the cellular and molecular processes taking place in MA+ cells, we performed a functional analysis of those genes. We have shown that evident MA subcellular localizations are: at the nuclear envelope; inside the nucleus; and diffusely in perinuclear cytoplasm. Postulating that MA may be a nuclear receptor agonist, we carried out docking simulations with PPARα and RORα ligand binding domains including prediction and molecular dynamics-based analysis of potential MA binding poses. Finally, we confirmed that MA treatment enhanced nuclear apoptosis in human PBMCs. The research presented here, in our view, indicates that: (i) MA activity is mediated by nuclear receptors; (ii) MA is a possible PPARα and/or RORα agonist; (iii) MA has an immunosuppressive effect; and (iv) MA induces apoptosis through the mitochondrial signaling pathway.
Collapse
Affiliation(s)
| | | | - Artem A Kiselev
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Andrey N Gorshkov
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| | | | - Kirill A Vasilyev
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| | | | | | - Alexey L Kovalenko
- Institute of Toxicology, Federal Medical-Biological Agency of Russia, St. Petersburg, Russia
| | - Andrey V Vasin
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
- Institute of Biomedical Systems and Botechnologies, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Saint Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| |
Collapse
|
121
|
Metabolic Remodelling: An Accomplice for New Therapeutic Strategies to Fight Lung Cancer. Antioxidants (Basel) 2019; 8:antiox8120603. [PMID: 31795465 PMCID: PMC6943435 DOI: 10.3390/antiox8120603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling is a hallmark of cancer, however little has been unravelled in its role in chemoresistance, which is a major hurdle to cancer control. Lung cancer is a leading cause of death by cancer, mainly due to the diagnosis at an advanced stage and to the development of resistance to therapy. Targeted therapeutic agents combined with comprehensive drugs are commonly used to treat lung cancer. However, resistance mechanisms are difficult to avoid. In this review, we will address some of those therapeutic regimens, resistance mechanisms that are eventually developed by lung cancer cells, metabolic alterations that have already been described in lung cancer and putative new therapeutic strategies, and the integration of conventional drugs and genetic and metabolic-targeted therapies. The oxidative stress is pivotal in this whole network. A better understanding of cancer cell metabolism and molecular adaptations underlying resistance mechanisms will provide clues to design new therapeutic strategies, including the combination of chemotherapeutic and targeted agents, considering metabolic intervenients. As cancer cells undergo a constant metabolic adaptive drift, therapeutic regimens must constantly adapt.
Collapse
|
122
|
Redox-Mediated Mechanism of Chemoresistance in Cancer Cells. Antioxidants (Basel) 2019; 8:antiox8100471. [PMID: 31658599 PMCID: PMC6826977 DOI: 10.3390/antiox8100471] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular reactive oxygen species (ROS) status is stabilized by a balance of ROS generation and elimination called redox homeostasis. ROS is increased by activation of endoplasmic reticulum stress, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family members and adenosine triphosphate (ATP) synthesis of mitochondria. Increased ROS is detoxified by superoxide dismutase, catalase, and peroxiredoxins. ROS has a role as a secondary messenger in signal transduction. Cancer cells induce fluctuations of redox homeostasis by variation of ROS regulated machinery, leading to increased tumorigenesis and chemoresistance. Redox-mediated mechanisms of chemoresistance include endoplasmic reticulum stress-mediated autophagy, increased cell cycle progression, and increased conversion to metastasis or cancer stem-like cells. This review discusses changes of the redox state in tumorigenesis and redox-mediated mechanisms involved in tolerance to chemotherapeutic drugs in cancer.
Collapse
|
123
|
Soberanes Y, Navarro RE, Inoue M, Velázquez-Contreras EF, Beltran Torres M, Lugo G, Sotelo-Mundo RR, Salazar-Medina AJ. Syntheses, Characterization, and Antioxidant Evaluation of Cu2+, Mn2+, and Fe3+ Complexes with a 14 Membered EDTA-Derived Macrocycle. Molecules 2019; 24:molecules24193556. [PMID: 31581425 PMCID: PMC6804257 DOI: 10.3390/molecules24193556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 11/19/2022] Open
Abstract
The Cu2+, Mn2+, and Fe3+ complexes of a 14 membered macrocycle were synthesized and their antioxidant capacities were evaluated against ABTS and DPPH radicals, with the objective of collecting insights into the biomimetic role of the central metal ions. The macrocycle, abbreviated as H2L14, is a derivative of EDTA cyclized with 1,4-diamine, and the moderately flexible macrocyclic frame permits the formation of [ML14·H2O] chelates with octahedral coordination geometries common among the metal ions. The metal complexes were characterized by electrospray-ionization mass spectrometry, Fourier transform infrared spectroscopy, and Raman and X-ray photoelectron spectroscopic methods, as well as thermogravimetric analysis; the octahedral coordination geometries with water coordination were optimized by DFT calculations. The antioxidant assays showed that [FeL14·H2O]+ was able to scavenge synthetic radicals with moderate capacity, whereas the other metal chelates did not show significant activity. The Raman spectrum of DPPH in solution suggests that interaction was operative between the Fe3+ chelate and the radical so as to cause scavenging capability. The nature of the central metal ions is a controlling factor for antioxidant capacity, as every metal chelate carries the same coordination geometry.
Collapse
Affiliation(s)
- Yedith Soberanes
- Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, Hermosillo Sonora 83000, Mexico.
- Laboratorio de Estructura Biomolecular, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo, Sonora 83304, Mexico.
| | - Rosa Elena Navarro
- Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, Hermosillo Sonora 83000, Mexico.
| | - Motomichi Inoue
- Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, Hermosillo Sonora 83000, Mexico.
| | | | - Melissa Beltran Torres
- Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, Hermosillo Sonora 83000, Mexico.
| | - Gustavo Lugo
- Departamento de Ingeniería Química, Universidad de Sonora, Hermosillo, Sonora 83000 Mexico.
| | - Rogerio R Sotelo-Mundo
- Laboratorio de Estructura Biomolecular, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo, Sonora 83304, Mexico.
| | - Alex J Salazar-Medina
- Cátedras CONACYT-Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, Hermosillo, Sonora 83000, Mexico.
| |
Collapse
|
124
|
Ramírez-Expósito MJ, Mayas MD, Carrera-González MP, Martínez-Martos JM. Gender Differences in the Antioxidant Response to Oxidative Stress in Experimental Brain Tumors. Curr Cancer Drug Targets 2019; 19:641-654. [DOI: 10.2174/1568009618666181018162549] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/13/2018] [Accepted: 09/15/2018] [Indexed: 01/16/2023]
Abstract
Background:Brain tumorigenesis is related to oxidative stress and a decreased response of antioxidant defense systems. As it is well known that gender differences exist in the incidence and survival rates of brain tumors, it is important to recognize and understand the ways in which their biology can differ.Objective:To analyze gender differences in redox status in animals with chemically-induced brain tumors.Methods:Oxidative stress parameters, non-enzyme and enzyme antioxidant defense systems are assayed in animals with brain tumors induced by transplacental N-ethyl-N-nitrosourea (ENU) administration. Both tissue and plasma were analyzed to know if key changes in redox imbalance involved in brain tumor development were reflected systemically and could be used as biomarkers of the disease.Results:Several oxidative stress parameters were modified in tumor tissue of male and female animals, changes that were not reflected at plasma level. Regarding antioxidant defense system, only glutathione (GSH) levels were decreased in both brain tumor tissue and plasma. Superoxide dismutase (SOD) and catalase (CAT) activities were decreased in brain tumor tissue of male and female animals, but plasma levels were only altered in male animals. However, different protein and mRNA expression patterns were found for both enzymes. On the contrary, glutathione peroxidase (GPx) activity showed increased levels in brain tumor tissue without gender differences, being protein and gene expression also increased in both males and female animals. However, these changes in GPx were not reflected at plasma level.Conclusion:We conclude that brain tumorigenesis was related to oxidative stress and changes in brain enzyme and non-enzyme antioxidant defense systems with gender differences, whereas plasma did not reflect the main redox changes that occur at the brain level.
Collapse
Affiliation(s)
| | - María Dolores Mayas
- Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| | | | | |
Collapse
|
125
|
da Fonseca TG, Abessa DMS, Bebianno MJ. Effects of mixtures of anticancer drugs in the benthic polychaete Nereis diversicolor. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:1180-1192. [PMID: 31252116 DOI: 10.1016/j.envpol.2019.05.095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 05/24/2023]
Abstract
The increasing consumption of anticancer drugs through single and/or combinatory chemotherapy worldwide raised concern regarding their toxicity burden in coastal zones. The toxicity of a mixture of three compounds involving the drugs cisplatin (CisPt), cyclophosphamide (CP) and tamoxifen (TAM) was determined on the marine polychaete Nereis diversicolor exposed to an increasing range of their concentrations, respectively: Mix A: 0.1 + 10 + 0.1 ng L-1; Mix B: 10 + 100 + 10 ng L-1; Mix C: 100 + 500 + 25 ng L-1; Mix D: 100 + 1000 + 100 ng L-1. Different endpoints were assessed, including disturbance in the burrowing behaviour, neurotoxicity (acetylcholinesterase - AChE activity), antioxidant enzymes (superoxide dismutase - SOD; catalase - CAT; selenium-dependent glutathione peroxidase - Se-GPx and total glutathione peroxidases T-GPx activities), biotransformation metabolism (glutathione-S-transferases - GST), lipid peroxidation (LPO) and genotoxicity (DNA damage). Biological effects of the mixtures of anticancer compounds on N. diversicolor were compared with previous studies about effects on the same biological model under single-drug exposure conducted with the same molecules. Regarding SOD activity, TAM showed an antagonist effect over CisPt and CP in mixtures C and D. In Mix D, there was a synergistic effect of TAM and CisPt that inhibited CAT activity and an additive interaction of CisPt and CP on the Phase II biotransformation enzyme. Drugs in Mix A also suppressed polychaetes' GST activity, although different from the respective single-drug responses, besides able to induce T-GPx activity, that was not sufficient to avoid oxidative damage and mid-grade DNA damage. Due to the absence of burrowing impairment in Mix A, mechanisms involved in neurotoxicity were other than the one driven by AChE alterations. At the intermediary concentrations (Mix B and C), only LPO occurred. Data from drugs individually may not predict the risks provided by mixtures.
Collapse
Affiliation(s)
- Tainá Garcia da Fonseca
- Centro de Investigação Marinha e Ambiental (CIMA), Universidade do Algarve, Campus de Gambelas, Faro, 8005-139, Portugal; NEPEA, Núcleo de Estudos em Poluição e Ecotoxicologia. Aquática, Universidade Estadual Paulista (UNESP), Campus do Litoral Paulista, São Vicente, SP, 11330-900, Brazil
| | - Denis M S Abessa
- NEPEA, Núcleo de Estudos em Poluição e Ecotoxicologia. Aquática, Universidade Estadual Paulista (UNESP), Campus do Litoral Paulista, São Vicente, SP, 11330-900, Brazil
| | - Maria João Bebianno
- Centro de Investigação Marinha e Ambiental (CIMA), Universidade do Algarve, Campus de Gambelas, Faro, 8005-139, Portugal.
| |
Collapse
|
126
|
Xiao J, Zhang G, Xu R, Chen H, Wang H, Tian G, Wang B, Yang C, Bai G, Zhang Z, Yang H, Zhong K, Zou D, Wu Z. A pH-responsive platform combining chemodynamic therapy with limotherapy for simultaneous bioimaging and synergistic cancer therapy. Biomaterials 2019; 216:119254. [DOI: 10.1016/j.biomaterials.2019.119254] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/27/2019] [Accepted: 06/05/2019] [Indexed: 01/11/2023]
|
127
|
Cocetta V, Ragazzi E, Montopoli M. Mitochondrial Involvement in Cisplatin Resistance. Int J Mol Sci 2019; 20:ijms20143384. [PMID: 31295873 PMCID: PMC6678541 DOI: 10.3390/ijms20143384] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/25/2022] Open
Abstract
Cisplatin is one of the worldwide anticancer drugs and, despite its toxicity and frequent recurrence of resistance phenomena, it still remains the only therapeutic option for several tumors. Circumventing cisplatin resistance remains, therefore, a major goal for clinical therapy and represents a challenge for scientific research. Recent studies have brought to light the fundamental role of mitochondria in onset, progression, and metastasis of cancer, as well as its importance in the resistance to chemotherapy. The aim of this review is to give an overview of the current knowledge about the implication of mitochondria in cisplatin resistance and on the recent development in this research field. Recent studies have highlighted the role of mitochondrial DNA alterations in onset of resistance phenomena, being related both to redox balance alterations and to signal crosstalk with the nucleus, allowing a rewiring of cell metabolism. Moreover, an important role of the mitochondrial dynamics in the adaptation mechanism of cancer cells to challenging environment has been revealed. Giving bioenergetic plasticity to tumor cells, mitochondria allow cells to evade death pathways in stressful conditions, including chemotherapy. So far, even if the central role of mitochondria is recognized, little is known about the specific mechanisms implicated in the resistance. Nevertheless, mitochondria appear to be promising pharmacological targets for overcoming cisplatin resistance, but further studies are necessary.
Collapse
Affiliation(s)
- Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Egidio Meneghetti 2, 35131 Padua, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Egidio Meneghetti 2, 35131 Padua, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Egidio Meneghetti 2, 35131 Padua, Italy.
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padua, Italy.
| |
Collapse
|
128
|
Gao J, Li T, Lu Z, Wang X, Zhao X, Ma Y. Proteomic Analyses of Mammary Glands Provide Insight into the Immunity and Metabolism Pathways Associated with Clinical Mastitis in Meat Sheep. Animals (Basel) 2019; 9:ani9060309. [PMID: 31159303 PMCID: PMC6617192 DOI: 10.3390/ani9060309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Clinical mastitis is one of the most common diseases in sheep and is of major economic concern due to treatment costs, inadequate lamb growth and premature eliminate of ewes. To preliminarily explore possible regulatory roles of proteins involved in the host-pathogen interactions during intramammary infection triggered by this disease in meat sheep, mammary tissues were harvested from sheep with healthy and clinical mastitis caused by natural infection, and the differentially expressed proteins were identified in an infected group when compared to a healthy group, using comparative proteomics based on two-dimensional electrophoresis. Further enrichment analyses indicated that most of the differentially expressed proteins mainly engaged in regulating immune responses and metabolisms. These findings offer candidate proteins for further studies on molecular mechanisms of host defense response and metabolism in sheep cases. Abstract Clinical mastitis is still an intractable problem for sheep breeding. The natural immunologic mechanisms of the mammary gland against infections are not yet understood. For a better understanding of the disease-associated proteins during clinical mastitis in meat sheep, we performed two-dimensional electrophoresis (2-DE)-based comparative proteomic analyses of mammary tissues, including from healthy mammary tissues (HMTs) and from mammary tissues with clinical mastitis (CMMTs). The 2-DE results showed that a total of 10 up-regulated and 16 down-regulated proteins were identified in CMMTs when compared to HMTs. Of these, Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analyses revealed that most proteins were associated with immune responses or metabolisms. The results of qRT-PCR and Western blot for randomly selected four differentially expressed proteins (DEPs) including superoxide dismutase [Mn] (SOD2), annexin A2 (ANAX2), keratin 10 (KRT10) and endoplasmic reticulum resident protein 29 (ERP29) showed that their expression trends were consistent with 2-DE results except ANXA2 mRNA levels. This is an initial report describing the 2-DE-based proteomics study of the meat sheep mammary gland with clinical mastitis caused by natural infection, which provides additional insight into the immune and metabolic mechanisms during sheep mastitis.
Collapse
Affiliation(s)
- Jianfeng Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Taotao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
- Sheep Breeding Biotechnology Engineering Laboratory of Gansu Province, Minqin 733300, China.
| | - Zengkui Lu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Xia Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China.
| | - Youji Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
- Sheep Breeding Biotechnology Engineering Laboratory of Gansu Province, Minqin 733300, China.
| |
Collapse
|
129
|
Solingapuram Sai KK, Bashetti N, Chen X, Norman S, Hines JW, Meka O, Kumar JVS, Devanathan S, Deep G, Furdui CM, Mintz A. Initial biological evaluations of 18F-KS1, a novel ascorbate derivative to image oxidative stress in cancer. EJNMMI Res 2019; 9:43. [PMID: 31101996 PMCID: PMC6525227 DOI: 10.1186/s13550-019-0513-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS)-induced oxidative stress damages many cellular components such as fatty acids, DNA, and proteins. This damage is implicated in many disease pathologies including cancer and neurodegenerative and cardiovascular diseases. Antioxidants like ascorbate (vitamin C, ascorbic acid) have been shown to protect against the deleterious effects of oxidative stress in patients with cancer. In contrast, other data indicate potential tumor-promoting activity of antioxidants, demonstrating a potential temporal benefit of ROS. However, quantifying real-time tumor ROS is currently not feasible, since there is no way to directly probe global tumor ROS. In order to study this ROS-induced damage and design novel therapeutics to prevent its sequelae, the quantitative nature of positron emission tomography (PET) can be harnessed to measure in vivo concentrations of ROS. Therefore, our goal is to develop a novel translational ascorbate-based probe to image ROS in cancer in vivo using noninvasive PET imaging of tumor tissue. The real-time evaluations of ROS state can prove critical in developing new therapies and stratifying patients to therapies that are affected by tumor ROS. METHODS We designed, synthesized, and characterized a novel ascorbate derivative (E)-5-(2-chloroethylidene)-3-((4-(2-fluoroethoxy)benzyl)oxy)-4-hydroxyfuran-2(5H)-one (KS1). We used KS1 in an in vitro ROS MitoSOX-based assay in two different head and neck squamous cancer cells (HNSCC) that express different ROS levels, with ascorbate as reference standard. We radiolabeled 18F-KS1 following 18F-based nucleophilic substitution reactions and determined in vitro reactivity and specificity of 18F-KS1 in HNSCC and prostate cancer (PCa) cells. MicroPET imaging and standard biodistribution studies of 18F-KS1 were performed in mice bearing PCa cells. To further demonstrate specificity, we performed microPET blocking experiments using nonradioactive KS1 as a blocker. RESULTS KS1 was synthesized and characterized using 1H NMR spectra. MitoSOX assay demonstrated good correlations between increasing concentrations of KS1 and ascorbate and increased reactivity in SCC-61 cells (with high ROS levels) versus rSCC-61cells (with low ROS levels). 18F-KS1 was radiolabeled with high radiochemical purity (> 94%) and specific activity (~ 100 GBq/μmol) at end of synthesis (EOS). Cell uptake of 18F-KS1 was high in both types of cancer cells, and the uptake was significantly blocked by nonradioactive KS1, and the ROS blocker, superoxide dismutase (SOD) demonstrating specificity. Furthermore, 18F-KS1 uptake was increased in PCa cells under hypoxic conditions, which have been shown to generate high ROS. Initial in vivo tumor uptake studies in PCa tumor-bearing mice demonstrated that 18F-KS1 specifically bound to tumor, which was significantly blocked (threefold) by pre-injecting unlabeled KS1. Furthermore, biodistribution studies in the same tumor-bearing mice showed high tumor to muscle (target to nontarget) ratios. CONCLUSION This work demonstrates the strong preliminary support of 18F-KS1, both in vitro and in vivo for imaging ROS in cancer. If successful, this work will provide a new paradigm to directly probe real-time oxidative stress levels in vivo. Our work could enhance precision medicine approaches to treat cancer, as well as neurodegenerative and cardiovascular diseases affected by ROS.
Collapse
Affiliation(s)
| | - Nagaraju Bashetti
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh 522502 India
| | - Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Skylar Norman
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Justin W. Hines
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Omsai Meka
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - J. V. Shanmukha Kumar
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh 522502 India
| | | | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032 USA
| |
Collapse
|
130
|
Elkholi R, Abraham-Enachescu I, Trotta AP, Rubio-Patiño C, Mohammed JN, Luna-Vargas MPA, Gelles JD, Kaminetsky JR, Serasinghe MN, Zou C, Ali S, McStay GP, Pfleger CM, Chipuk JE. MDM2 Integrates Cellular Respiration and Apoptotic Signaling through NDUFS1 and the Mitochondrial Network. Mol Cell 2019; 74:452-465.e7. [PMID: 30879903 PMCID: PMC6499641 DOI: 10.1016/j.molcel.2019.02.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 11/30/2018] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
Signaling diversity and subsequent complexity in higher eukaryotes is partially explained by one gene encoding a polypeptide with multiple biochemical functions in different cellular contexts. For example, mouse double minute 2 (MDM2) is functionally characterized as both an oncogene and a tumor suppressor, yet this dual classification confounds the cell biology and clinical literatures. Identified via complementary biochemical, organellar, and cellular approaches, we report that MDM2 negatively regulates NADH:ubiquinone oxidoreductase 75 kDa Fe-S protein 1 (NDUFS1), leading to decreased mitochondrial respiration, marked oxidative stress, and commitment to the mitochondrial pathway of apoptosis. MDM2 directly binds and sequesters NDUFS1, preventing its mitochondrial localization and ultimately causing complex I and supercomplex destabilization and inefficiency of oxidative phosphorylation. The MDM2 amino-terminal region is sufficient to bind NDUFS1, alter supercomplex assembly, and induce apoptosis. Finally, this pathway is independent of p53, and several mitochondrial phenotypes are observed in Drosophila and murine models expressing transgenic Mdm2.
Collapse
Affiliation(s)
- Rana Elkholi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Ioana Abraham-Enachescu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew P Trotta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Camila Rubio-Patiño
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jarvier N Mohammed
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Mark P A Luna-Vargas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Joshua R Kaminetsky
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Madhavika N Serasinghe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Cindy Zou
- Department of Life Sciences, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA
| | - Sumaira Ali
- Department of Life Sciences, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA
| | - Gavin P McStay
- Department of Life Sciences, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA
| | - Cathie M Pfleger
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
131
|
Ding F, Sun K, Sun N, Jiang Q, Cao M, Wu Z. iTRAQ-based proteomics reveals SOD2 as a potential salivary biomarker in liver cancer. Int J Biol Markers 2019; 34:221-231. [PMID: 31041878 DOI: 10.1177/1724600819841619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Salivary proteomic analysis has been extensively used in a wide range of cancer, but not in hepatocellular carcinoma. The aim of this study was to identify potential salivary biomarkers for hepatocellular carcinoma clinical screening. METHODS In this study, we performed isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics analysis to detect differentially expressed proteins between saliva samples from 15 hepatocellular carcinoma patients and 15 healthy controls. Enzyme-linked immunosorbent assay (ELISA) verification was undertaken in saliva samples from 14 hepatocellular carcinoma patients and 14 healthy controls. RESULTS Overall, 133 proteins with significant differential expression level (ratio > 1.5 or < 0.67) were detected. Using bioinformatic analysis, two candidate proteins were selected and subsequently verified by ELISA. The increased expression of superoxide dismutase 2, mitochondrial (SOD2) in hepatocellular carcinoma patients was confirmed by ELISA, with an area under the curve value of 0.9082. CONCLUSIONS iTRAQ-based quantitative proteomics revealed that SOD2 might serve as a potential salivary biomarker for hepatocellular carcinoma detection. Our results indicated that a noninvasive and inexpensive salivary test might be established for hepatocellular carcinoma detection.
Collapse
Affiliation(s)
- Feng Ding
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Kehuan Sun
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Ningning Sun
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Qianqian Jiang
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Meiqun Cao
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Zhengzhi Wu
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
132
|
Gomez M, Germain D. Cross talk between SOD1 and the mitochondrial UPR in cancer and neurodegeneration. Mol Cell Neurosci 2019; 98:12-18. [PMID: 31028834 DOI: 10.1016/j.mcn.2019.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 01/23/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is rapidly gaining attention. While the CHOP (ATF4/5) axis of the UPRmt was the first to be described, other axes have subsequently been reported. Validation of this complex pathway in C. elegans has been extensively studied. However, validation of the UPRmt in mouse models of disease known to implicate mitochondrial reprogramming or dysfunction, such as cancer and neurodegeneration, respectively, is only beginning to emerge. This review summarizes recent findings and highlights the major role of the superoxide dismutase SOD1 in the communication between the mitochondria and the nucleus in these settings. While SOD1 has mostly been studied in the context of familial amyotrophic lateral sclerosis (fALS), recent studies suggest that SOD1 may be a potentially important mediator of the UPRmt and converge to emphasize an increasingly vital role of SOD1 as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Maria Gomez
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, New York, 10029, NY, USA
| | - Doris Germain
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, New York, 10029, NY, USA.
| |
Collapse
|
133
|
Dromparis P, Aboelnazar NS, Wagner S, Himmat S, White CW, Hatami S, Luc JGY, Rotich S, Freed DH, Nagendran J, Mengel M, Adam BA. Ex vivo perfusion induces a time- and perfusate-dependent molecular repair response in explanted porcine lungs. Am J Transplant 2019; 19:1024-1036. [PMID: 30230229 DOI: 10.1111/ajt.15123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 01/25/2023]
Abstract
Ex vivo lung perfusion (EVLP) shows promise in ameliorating pretransplant acute lung injury (ALI) and expanding the donor organ pool, but the mechanisms of ex vivo repair remain poorly understood. We aimed to assess the utility of gene expression for characterizing ALI during EVLP. One hundred sixty-nine porcine lung samples were collected in vivo (n = 25), after 0 (n = 11) and 12 (n = 11) hours of cold static preservation (CSP), and after 0 (n = 57), 6 (n = 8), and 12 (n = 57) hours of EVLP, utilizing various ventilation and perfusate strategies. The expression of 53 previously described ALI-related genes was measured and correlated with function and histology. Twenty-eight genes were significantly upregulated and 6 genes downregulated after 12 hours of EVLP. Aggregate gene sets demonstrated differential expression with EVLP (P < .001) but not CSP. Upregulated 28-gene set expression peaked after 6 hours of EVLP, whereas downregulated 6-gene set expression continued to decline after 12 hours. Cellular perfusates demonstrated a greater reduction in downregulated 6-gene set expression vs acellular perfusate (P < .038). Gene set expression correlated with relevant functional and histologic parameters, including P/F ratio (P < .001) and interstitial inflammation (P < .005). Further studies with posttransplant results are warranted to evaluate the clinical significance of this novel molecular approach for assessing organ quality during EVLP.
Collapse
Affiliation(s)
- Peter Dromparis
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Nader S Aboelnazar
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Siegfried Wagner
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Sayed Himmat
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher W White
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Sanaz Hatami
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jessica G Y Luc
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Silas Rotich
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jayan Nagendran
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
134
|
Li J, Lei J, He L, Fan X, Yi F, Zhang W. Evaluation and Monitoring of Superoxide Dismutase (SOD) Activity and its Clinical Significance in Gastric Cancer: A Systematic Review and Meta-Analysis. Med Sci Monit 2019; 25:2032-2042. [PMID: 30886134 PMCID: PMC6436205 DOI: 10.12659/msm.913375] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background This systematic review of the literature and meta-analysis aimed to review the evaluation and monitoring of superoxide dismutase (SOD) activity and its clinical significance in gastric cancer. Material/Methods Systematic review involved searching the PubMed, Embase, Ovid, and the China National Knowledge Infrastructure (CNKI) databases. Search terms included ‘superoxide dismutase,’ and ‘gastric cancer.’ Studies that included measurements of SOD activity in peripheral blood samples in patients with SOD activity compared with healthy controls. The study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Results Ten controlled clinical studies were identified that included six studies that measured SOD in serum, three in erythrocytes, and one study that measured SOD on whole blood. Meta-analysis, using the standardized mean difference (SMD) and the 95% confidence interval (CI), showed that patients with gastric cancer had significantly decreased SOD activity when compared with the healthy controls (SMD, −0.840; 95% CI, −1.463 to −0.218; p=0.008). Subgroup analysis was conducted on SOD distribution in the blood (erythrocyte: SMD, −1.773; 95% CI, −2.504 to −1.042; p=0.000) (serum SMD, −0.322; 95% CI, −1.006−0.361; p=0.355) (whole blood: SMD, −1.251; 95% CI, −1.731 to −0.771; p=0.000) and for male subjects (SMD, −2.090; 95% CI, −2.725 to −1.456; p<0.001). Conclusions Meta-analysis showed that SOD measurements from blood samples, especially in erythrocytes, had potential as a diagnostic and monitoring parameter in patients with gastric cancer.
Collapse
Affiliation(s)
- Jine Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland).,Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Jun Lei
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Liyun He
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland).,Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Xiude Fan
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Fengming Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
135
|
Caglayan A, Katlan DC, Tuncer ZS, Yüce K. Evaluation of trace elements associated with antioxidant enzymes in blood of primary epithelial ovarian cancer patients. J Trace Elem Med Biol 2019; 52:254-262. [PMID: 30732891 DOI: 10.1016/j.jtemb.2019.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/18/2018] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
Epithelial ovarian cancer (EOC) has been associated with oxidative stress (OS) due to epithelial inflammation which makes ovaries more vulnerable to the deleterious effects of reactive oxygen species (ROS). However, antioxidant enzymes (AOEs) such as manganese-superoxide dismutase (Mn-SOD), copper,zinc-superoxide dismutase (Cu,Zn-SOD) and glutathione peroxidase (GPx1) protect cells against the biological damage of ROS-induced OS and support cancer prevention by maintaining normal cell cycle progression, inhibiting proliferation, tumor invasion, angiogenesis, inflammation or inducing apoptosis. In the present study, we aimed to measure the levels of trace elements [manganese (Mn), copper (Cu), zinc (Zn) and selenium (Se)] which are structurally and/or functionally associated with the AOEs by inductively coupled plasma/mass-spectrometry (ICP/MS) in blood samples of patients with EOC (M, n = 26) and compare the data with healthy subjects (C, n = 46). Serous EOC (M1, n = 18) data were also evaluated according to the tumor grading [well or moderately well differentiated (G 1-2) vs. poorly differentiated or undifferentiated (G3)] and staging of disease [stage I-II (SI-II) vs. stage III (SIII)]. We obtained; i) The Mn and Se levels of M were significantly lower than C, ii) only Mn levels were changed [(G3(Mn) < G 1-2 (Mn)] in M1, iii) significant correlations were observed between [Cu and Zn levels (r = 0.701, p = 0.036) in G 1-2 and (r = 0.686, p = 0.041) in G3; Cu and Se levels (r = 0.960, p = 0.000) in G3; Mn levels and Mn-SOD expression (r = 0.551, p = 0.006) in M, (r = 0.857, p = 0.007) in G 1-2 and (r = 0.690, p = 0.056) in G3; Se levels and erythrocyte GPx1 activity (r = 0.660, p = 0.053) in G 1-2 ; Se levels and erythrocyte Cu,Zn-SOD activity (r = 0.693, p = 0.038) in G3]. The study revealed that trace elements, particularly low Mn and Se levels along with high Cu/Se ratios might be of value in all histologic subtypes of EOC. Although Mn level was important in terms of discriminating tumor grades, positive correlation between Cu-Se levels was also remarkable in patients with G 1-2 tumors of M1. Moreover, high erythrocyte Cu/Se ratios might be a favourable marker for EOC.
Collapse
Affiliation(s)
- Aydan Caglayan
- Hacettepe University, Faculty of Pharmacy, Department of Toxicology, Ankara 06100, Turkey.
| | - Doruk Cevdi Katlan
- İstanbul Education and Research Hospital, Obstetrics and Gynecology, İstanbul 34020, Turkey.
| | - Zafer Selcuk Tuncer
- Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Ankara 06100, Turkey.
| | - Kunter Yüce
- Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Ankara 06100, Turkey.
| |
Collapse
|
136
|
Zhang X, Ye L, Kang Z, Zou J, Zhang X, Li X. Mycorrhization of Quercus acutissima with Chinese black truffle significantly altered the host physiology and root-associated microbiomes. PeerJ 2019; 7:e6421. [PMID: 30805248 PMCID: PMC6383558 DOI: 10.7717/peerj.6421] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/07/2019] [Indexed: 01/16/2023] Open
Abstract
Background Our aim was to explore how the ectomycorrhizae of an indigenous tree,Quercus acutissima, with a commercial truffle, Chinese black truffle (Tuber indicum), affects the host plant physiology and shapes the associated microbial communities in the surrounding environment during the early stage of symbiosis. Methods To achieve this, changes in root morphology and microscopic characteristics, plant physiology indices, and the rhizosphere soil properties were investigated when six-month-old ectomycorrhizae were synthesized. Meanwhile, next-generation sequencing technology was used to analyze the bacterial and fungal communities in the root endosphere and rhizosphere soil inoculated with T. indicum or not. Results The results showed that colonization by T. indicum significantly improved the activity of superoxide dismutase in roots but significantly decreased the root activity. The biomass, leaf chlorophyll content and root peroxidase activity did not obviously differ. Ectomycorrhization of Q. acutissima with T. indicum affected the characteristics of the rhizosphere soil, improving the content of organic matter, total nitrogen, total phosphorus and available nitrogen. The bacterial and fungal community composition in the root endosphere and rhizosphere soil was altered by T. indicum colonization, as was the community richness and diversity. The dominant bacteria in all the samples were Proteobacteria and Actinobacteria, and the dominant fungi were Eukaryota_norank, Ascomycota, and Mucoromycota. Some bacterial communities, such as Streptomyces, SM1A02, and Rhizomicrobium were more abundant in the ectomycorrhizae or ectomycorrhizosphere soil. Tuber was the second-most abundant fungal genus, and Fusarium was present at lower amounts in the inoculated samples. Discussion Overall, the symbiotic relationship between Q. acutissima and T. indicum had an obvious effect on host plant physiology, soil properties, and microbial community composition in the root endosphere and rhizosphere soil, which could improve our understanding of the symbiotic relationship between Q. acutissima and T. indicum, and may contribute to the cultivation of truffle.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Soil and Fertilizer Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China.,Department of Microbiology, College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Lei Ye
- Soil and Fertilizer Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Zongjing Kang
- Soil and Fertilizer Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China.,Department of Microbiology, College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Jie Zou
- Soil and Fertilizer Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China.,Department of Microbiology, College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Xiaoping Zhang
- Department of Microbiology, College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Xiaolin Li
- Soil and Fertilizer Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
137
|
Li X, Chen Y, Zhao J, Shi J, Wang M, Qiu S, Hu Y, Xu Y, Cui Y, Liu C, Liu C. The Specific Inhibition of SOD1 Selectively Promotes Apoptosis of Cancer Cells via Regulation of the ROS Signaling Network. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9706792. [PMID: 30911355 PMCID: PMC6398008 DOI: 10.1155/2019/9706792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 12/02/2018] [Indexed: 12/17/2022]
Abstract
Multiple signaling pathways including ERK, PI3K-Akt, and NF-κB, which are essential for onset and development of cancer, can be activated by intracellularly sustained high levels of H2O2 provided by elevated activity and expression of copper/zinc superoxide dismutase (SOD1) that catalyzes the dismutation of O2 •- into H2O2. Here, tests performed by the utilization of our designed specific SOD1 inhibitor LD100 on cancer and normal cells reveal that the signaling pathways and their crosstalk to support cancer cell growth are repressed, but the signaling pathways to promote cancer cell cycle arrest and apoptosis are stimulated by specific SOD1 inhibition-mediated ROS changes. These regulated pathways constitute an ROS signaling network that determines the fate of cancer cells. This ROS signaling network is also regulated in SOD1 knockdown cells. These findings might facilitate disclosure of action mechanisms by copper-chelating anticancer agents and design of SOD1-targeting and ROS signaling pathway-interfering anticancer small molecules.
Collapse
Affiliation(s)
- Xiang Li
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yuanyuan Chen
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Jidong Zhao
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Jiayuan Shi
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Mingfang Wang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Shuang Qiu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yinghui Hu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yulin Xu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Yanfang Cui
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Chunrong Liu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| | - Changlin Liu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, School of Chemistry, Central China Normal University, Wuhan, 430079 Hubei, China
| |
Collapse
|
138
|
Mitochondrial superoxide dismutase 2 mediates γ-irradiation-induced cancer cell invasion. Exp Mol Med 2019; 51:1-10. [PMID: 30755594 PMCID: PMC6372678 DOI: 10.1038/s12276-019-0207-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/04/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022] Open
Abstract
Sublethal doses of γ-rays promote cancer cell invasion by stimulating a signaling pathway that sequentially involves p53, sulfatase 2 (SULF2), β-catenin, interleukin-6 (IL-6), signal transducer and activator of transcription 3 (STAT3), and Bcl-XL. Given that Bcl-XL can increase O2•− production by stimulating respiratory complex I, the possible role of mitochondrial reactive oxygen species (ROS) in γ-irradiation-induced cell invasion was investigated. Indeed, γ-irradiation promoted cell invasion by increasing mitochondrial ROS levels, which was prevented by metformin, an inhibitor of complex I. γ-Irradiation-stimulated STAT3 increased the expression of superoxide dismutase 2 (SOD2), a mitochondrial enzyme that catalyzes the conversion of O2•− to hydrogen peroxide (H2O2). In contrast to O2•−, H2O2 functions as a signaling molecule. γ-Irradiation consistently stimulated the Src-dependent invasion pathway in a manner dependent on both complex I and SOD2. SOD2 was also essential for the invasion of un-irradiated cancer cells induced by upregulation of Bcl-XL, an intracellular oncogene, or extracellular factors, such as SULF2 and IL-6. Overall, these data suggested that SOD2 is critical for the malignant effects of radiotherapy and tumor progression through diverse endogenous factors. A drug usually used to treat type 2 diabetes may also help to prevent cancer relapse following radiotherapy, which is commonly used to kill cancer cells. However, any tumor cells that survive radiation are highly invasive, sometimes causing tumors to spread. Hong-Duck Um and co-workers at the Korea Institute of Radiological & Medical Sciences in Seoul, South Korea, noticed that the surviving cells often showed higher levels of a key enzyme, superoxide dismutase 2 (SOD2), which is involved in energy production in the cellular powerhouse, the mitochondria. Artificially increasing levels of SOD2, without radiation, made cells more invasive. Treatment with metformin, which prevents production of the molecule that SOD2 acts on, prevented cells from becoming invasive. SOD2 has been implicated in many cancers, and is therefore a very promising therapeutic target.
Collapse
|
139
|
Ramírez-Expósito MJ, Martínez-Martos JM. The Delicate Equilibrium between Oxidants and Antioxidants in Brain Glioma. Curr Neuropharmacol 2019; 17:342-351. [PMID: 29512467 PMCID: PMC6482474 DOI: 10.2174/1570159x16666180302120925] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/03/2018] [Accepted: 02/02/2018] [Indexed: 11/22/2022] Open
Abstract
Gliomas are the most frequent brain tumors in the adult population and unfortunately the adjuvant therapies are not effective. Brain tumorigenesis has been related both to the increased levels of free radicals as inductors of severe damages in healthy cells, but also with the reduced response of endogenous enzyme and non-enzymatic antioxidant defenses. In turn, both processes induce the change to malignant cells. In this review, we analyzed the role of the imbalance between free radicals production and antioxidant mechanism in the development and progression of gliomas but also the influence of redox status on the two major distinctive forms of programmed cell death related to cancer: apoptosis and autophagy. These data may be the reference to the development of new pharmacological options based on redox microenvironment for glioma treatment.
Collapse
Affiliation(s)
- María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS-1039; Department of Health Sciences, Faculty of Health Sciences; University of Jaén, Campus Universitario Las Lagunillas, Jaén, Spain
| | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS-1039; Department of Health Sciences, Faculty of Health Sciences; University of Jaén, Campus Universitario Las Lagunillas, Jaén, Spain
| |
Collapse
|
140
|
Dajani S, Saripalli A, Sharma-Walia N. Water transport proteins-aquaporins (AQPs) in cancer biology. Oncotarget 2018; 9:36392-36405. [PMID: 30555637 PMCID: PMC6284741 DOI: 10.18632/oncotarget.26351] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/22/2018] [Indexed: 02/06/2023] Open
Abstract
As highly conserved ubiquitous proteins, aquaporins (AQPs) play an imperative role in the development and progression of cancer. By trafficking water and other small molecules, AQPs play a vital role in preserving the cellular environment. Due to their critical role in cell stability and integrity, it would make sense that AQPs are involved in cancer progression. When AQPs alter the cellular environment, there may be several downstream effects such as alterations in cellular osmolality, volume, ionic composition, and signaling pathways. Changes in the intracellular levels of certain molecules serving as second messengers are synchronized by AQPs. Thus AQPs regulate numerous downstream effector signaling molecules that promote cancer development and progression. In numerous cancer types, AQP expression has shown a correlation with tumor stage and prognosis. Furthermore, AQPs assist in angiogenic and oxidative stress related damaging processes critical for cancer progression. This indicates that AQP proteins may be a viable therapeutic target or biomarker of cancer prognosis.
Collapse
Affiliation(s)
- Salah Dajani
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Anand Saripalli
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Neelam Sharma-Walia
- H.M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
141
|
Tsang CK, Zheng XFS. A balancing act: mTOR integrates nutrient signals to regulate redox-dependent growth and survival through SOD1. Mol Cell Oncol 2018; 5:e1488372. [PMID: 30263944 PMCID: PMC6154846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/09/2018] [Indexed: 06/08/2023]
Abstract
Maintaining cellular redox is critical for growth, metabolism and survival in response to changing environments. How nutrients regulate this process is a long-standing fundamental question in cell biology. Our recent study revealed a conserved mechanism by which eukaryotes, particularly cancer cells, couple nutrient signaling to dynamically regulate redox homeostasis. Abbreviations: ATP: adenosine triphosphate; Ala: alanine; C6H12O6: glucose; OH-: hydroxyl radical; Glu: glutamate; mRNA: messenger RNA; mTOR: mechanistic/mammalian target of rapamycin; OXYPHOS: oxidative phosphorylation; Ser: serine; ROS: reactive oxygen species; O2 -: superoxide; SOD1: superoxide dismutase 1; Thr: threonine.
Collapse
Affiliation(s)
- Chi Kwan Tsang
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ USA
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ USA
- Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong China
| | - X. F. Steven Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ USA
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ USA
| |
Collapse
|
142
|
Recent trends in electrochemical biosensors of superoxide dismutases. Biosens Bioelectron 2018; 116:89-99. [DOI: 10.1016/j.bios.2018.05.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 01/16/2023]
|
143
|
Ježek P, Holendová B, Garlid KD, Jabůrek M. Mitochondrial Uncoupling Proteins: Subtle Regulators of Cellular Redox Signaling. Antioxid Redox Signal 2018; 29:667-714. [PMID: 29351723 PMCID: PMC6071544 DOI: 10.1089/ars.2017.7225] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Mitochondria are the energetic, metabolic, redox, and information signaling centers of the cell. Substrate pressure, mitochondrial network dynamics, and cristae morphology state are integrated by the protonmotive force Δp or its potential component, ΔΨ, which are attenuated by proton backflux into the matrix, termed uncoupling. The mitochondrial uncoupling proteins (UCP1-5) play an eminent role in the regulation of each of the mentioned aspects, being involved in numerous physiological events including redox signaling. Recent Advances: UCP2 structure, including purine nucleotide and fatty acid (FA) binding sites, strongly support the FA cycling mechanism: UCP2 expels FA anions, whereas uncoupling is achieved by the membrane backflux of protonated FA. Nascent FAs, cleaved by phospholipases, are preferential. The resulting Δp dissipation decreases superoxide formation dependent on Δp. UCP-mediated antioxidant protection and its impairment are expected to play a major role in cell physiology and pathology. Moreover, UCP2-mediated aspartate, oxaloacetate, and malate antiport with phosphate is expected to alter metabolism of cancer cells. CRITICAL ISSUES A wide range of UCP antioxidant effects and participations in redox signaling have been reported; however, mechanisms of UCP activation are still debated. Switching off/on the UCP2 protonophoretic function might serve as redox signaling either by employing/releasing the extra capacity of cell antioxidant systems or by directly increasing/decreasing mitochondrial superoxide sources. Rapid UCP2 degradation, FA levels, elevation of purine nucleotides, decreased Mg2+, or increased pyruvate accumulation may initiate UCP-mediated redox signaling. FUTURE DIRECTIONS Issues such as UCP2 participation in glucose sensing, neuronal (synaptic) function, and immune cell activation should be elucidated. Antioxid. Redox Signal. 29, 667-714.
Collapse
Affiliation(s)
- Petr Ježek
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| | - Blanka Holendová
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| | - Keith D Garlid
- 2 UCLA Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Martin Jabůrek
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| |
Collapse
|
144
|
Lee MY, Wu MF, Cherng SH, Chiu LY, Yang TY, Sheu GT. Tissue transglutaminase 2 expression is epigenetically regulated in human lung cancer cells and prevents reactive oxygen species-induced apoptosis. Cancer Manag Res 2018; 10:2835-2848. [PMID: 30197536 PMCID: PMC6112806 DOI: 10.2147/cmar.s155582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Tissue transglutaminase 2 (TG2) is a stress-regulated protein and associated with cancer cell survival. However, the effects of TG2 expression in human non-small-cell lung cancer (NSCLC) cells on reactive oxygen species (ROS) production and redox homeostasis have not been fully elucidated. Materials and methods We investigated the TG2 expression and activity in A549, H1299, H1355, and H460 lung cancer cells by Western blots and quantitative polymerase chain reaction assay. The enzyme-linked immunosorbent assay was used for transglutaminase activity. The epigenetic expression was characterized with histone deacetylase inhibitor trichostatin A and DNA methyltransferase inhibitor 5-Aza treatment. TG2 expression was inhibited by siRNA transfection and the intracellular calcium was measured by Flow-3AM assay, apoptosis was analyzed by Annexin V/propidium iodide assay, and intracellular ROS was detected by fluorescence-activated cell sorting analysis. The ROS scavenger N-acetyl-L-cysteine (NAC) was applied to reduce TG2-knockdown-induced oxidative stress. Results Only A549 cells expressing high levels of TG2 correlated with high TG2 activity. The expression of TG2 can be regulated by epigenetic regulation in A549, H1299, and H1355 cells. The data also show that TG2 reduction induces apoptosis in A549 and H1299 cells. Furthermore, increased intracellular ROS and calcium levels were both detected in TG2-reduced cells. Moreover, endoplasmic reticulum stress inhibitor (salubrinal) and antioxidant NAC were able to reduce ROS and calcium levels to recover cell viability. Interestingly, the extrinsic and intrinsic apoptosis pathways were activated with a p53 independence upon TG2 reduction. TG2 reduction not only attenuated AKT activation but also reduced superoxide dismutase 2 (SOD2) expression. Exogenous NAC partially recovered SOD2 expression, indicating that mitochondrial-mediated apoptosis accounts for a part of but not all of the TG2-reduction-related death. Conclusion TG2 plays a protection role in NSCLC cell lines. Regardless of the endogenous level of TG2 and p53 status, reduction of TG2 may result in oxidative stress that induces apop-tosis. Therefore, target TG2 expression represents a logical strategy for NSCLC management.
Collapse
Affiliation(s)
- Ming-Yang Lee
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Chiayi City, Taiwan.,Graduate Institute of Natural Healing Science, Nanhua University, Chiayi City, Taiwan
| | - Ming-Fang Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Divisions of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan,
| | - Shur-Hueih Cherng
- Department of Biotechnology, Hung Kuang University, Taichung, Taiwan
| | - Ling-Yen Chiu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Gwo-Tarng Sheu
- Divisions of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, .,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, .,Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan,
| |
Collapse
|
145
|
Wert KJ, Velez G, Cross MR, Wagner BA, Teoh-Fitzgerald ML, Buettner GR, McAnany JJ, Olivier A, Tsang SH, Harper MM, Domann FE, Bassuk AG, Mahajan VB. Extracellular superoxide dismutase (SOD3) regulates oxidative stress at the vitreoretinal interface. Free Radic Biol Med 2018; 124:408-419. [PMID: 29940351 PMCID: PMC6233711 DOI: 10.1016/j.freeradbiomed.2018.06.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
Oxidative stress is a pathogenic feature in vitreoretinal disease. However, the ability of the inner retina to manage metabolic waste and oxidative stress is unknown. Proteomic analysis of antioxidants in the human vitreous, the extracellular matrix opposing the inner retina, identified superoxide dismutase-3 (SOD3) that localized to a unique matrix structure in the vitreous base and cortex. To determine the role of SOD3, Sod3-/- mice underwent histological and clinical phenotyping. Although the eyes were structurally normal, at the vitreoretinal interface Sod3-/- mice demonstrated higher levels of 3-nitrotyrosine, a key marker of oxidative stress. Pattern electroretinography also showed physiological signaling abnormalities within the inner retina. Vitreous biopsies and epiretinal membranes collected from patients with diabetic vitreoretinopathy (DVR) and a mouse model of DVR showed significantly higher levels of nitrates and/or 3-nitrotyrosine oxidative stress biomarkers suggestive of SOD3 dysfunction. This study analyzes the molecular pathways that regulate oxidative stress in human vitreous substructures. The absence or dysregulation of the SOD3 antioxidant at the vitreous base and cortex results in increased oxidative stress and tissue damage to the inner retina, which may underlie DVR pathogenesis and other vitreoretinal diseases.
Collapse
Affiliation(s)
- Katherine J Wert
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States
| | - Gabriel Velez
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States
| | - Madeline R Cross
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Brett A Wagner
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Melissa L Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Garry R Buettner
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - J Jason McAnany
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, IL, United States
| | - Alicia Olivier
- Division of Comparative Pathology, Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stephen H Tsang
- Bernard and Shirlee Brown Glaucoma Laboratory and Barbara & Donald Jonas Laboratory of Regenerative Medicine, Columbia University, New York, NY, United States; Edward S. Harkness Eye Institute, Columbia University, New York, NY, United States; Departments of Ophthalmology, Pathology & Cell Biology, and Institute of Human Nutrition, Columbia University, New York, NY, United States
| | - Matthew M Harper
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States; Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, United States; Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, United States
| | - Frederick E Domann
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Vinit B Mahajan
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States; Palo Alto Veterans Administration, Palo Alto, CA, United States.
| |
Collapse
|
146
|
Zhang Y, Xu Z, Ding J, Tan C, Hu W, Li Y, Huang W, Xu Y. HZ08 suppresses RelB-activated MnSOD expression and enhances Radiosensitivity of prostate Cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:174. [PMID: 30053873 PMCID: PMC6062957 DOI: 10.1186/s13046-018-0849-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/17/2018] [Indexed: 12/18/2022]
Abstract
Background The development of radioresistance is one of main causes for therapeutic failure of prostate cancer (PCa). The present study aims to investigate the function and the related mechanism by which HZ08 sensitizes radiotherapeutic efficiency to treat aggressive PCa cells. Methods PCa cells were pretreated with HZ08 (6,7-dimethoxy-1-(3,4-dimethoxy) benzyl-2-(N-n-octyl-N′-cyano) guanyl-1,2,3,4-tetrahydroisoquinoline) and followed by ionizing radiation (IR) treatment. Cytotoxicity in the treated cells was analyzed to assess the radiosensitization capacity of HZ08 by flow cytometry, MTT and colony survival assays. The cellular levels of reactive oxygen species (ROS) and oxygen consumption rates (OCR) were measured using specific ROS detection probes and a Seahorse XF96 Analyzer, respectively. RelB binding to the NF-κB intronic enhancer region of the human SOD2 gene was determined using a ChIP assay. The levels of phosphorylation of PI3K, Akt and IKKα were quantified and further confirmed using a PI3K inhibitor. Finally, the synergistic effect of HZ08 on radiosensitization of PCa cells was validated using a mouse xenograft tumor model. Results HZ08 enhanced radiosensitivity of PCa cells through increasing ROS and declining mitochondrial respiration due to suppression of mitochondrial antioxidant enzyme MnSOD. Mechanistically, HZ08 appeared to inhibit PI3K/Akt/IKKα signaling axis, resulting in transcriptional repression of MnSOD expression by preventing RelB nuclear translocation. Conclusions HZ08 can serve as a useful radiosensitizing agent to improve radiotherapy for treating aggressive PCa cells with high level of constitutive RelB. The present study suggests a promising approach for enhancing radiotherapeutic efficiency to treat advanced PCa by inhibiting antioxidant defense function. Electronic supplementary material The online version of this article (10.1186/s13046-018-0849-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanyan Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China
| | - Zhi Xu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China
| | - Jiaji Ding
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China
| | - Chunli Tan
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China
| | - Weizi Hu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Wenlong Huang
- Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yong Xu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, People's Republic of China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
147
|
Zhang HL, Liu CY, Ma W, Huang L, Li CJ, Li CS, Zhang ZW. Identification of differentially expressed proteins in the gastric mucosal atypical hyperplasia tissue microenvironment. Oncol Lett 2018; 16:2355-2365. [PMID: 30008939 PMCID: PMC6036401 DOI: 10.3892/ol.2018.8941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 04/05/2018] [Indexed: 11/06/2022] Open
Abstract
In the present study, the interaction of proteins in the microenvironment of gastric mucosal atypical hyperplasia was analyzed. The stromata of normal gastric mucosa (NGM) and gastric mucosal atypical hyperplasia (GMAH) tissues were purified with laser capture microdissection (LCM). The differentially expressed GMAH proteins of the NGM and GMAH tissues were identified by quantitative proteomic techniques with isotope labeling. The cross-talk between differentially expressed proteins in NGM and GMAH tissues was then analyzed by bioinformatics. There were 165 differentially expressed proteins identified from the stromata of NGM and GMAH tissues. Among them, 99 proteins were upregulated and 66 were downregulated in GMAH tissue. The present study demonstrated that these proteins in gastric mucosal atypical hyperplasia were involved in cancer-associated signaling pathways, including the p53, mitogen-activated protein kinase (MAPK), cell cycle and apoptosis signaling pathways, and were involved in cellular growth, cellular proliferation, apoptosis and the humoral immune response. The results of the present study suggest that the 165 differentially expressed proteins, including S100 calcium-binding protein A6 (S100A6) and superoxide dismutase 3 (SOD3) in the microenvironment of gastric mucosal atypical hyperplasia, are involved in the p53, MAPK, cell cycle and apoptosis signaling pathways, and serve a function in the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- He-Liang Zhang
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Medical College, University of South China, Hengyang, Hunan 421001, P.R. China.,Medical Company, Troops 66028 of People's Liberation Army, Chengde, Hebei 067000, P.R. China
| | - Chong-Yuan Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Ma
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lin Huang
- Department of Pediatrics, Shaoyang Medical School, Shaoyang, Hunan 422000, P.R. China
| | - Chang-Jian Li
- Clinical Medicine Undergraduate Program, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Cheng-Song Li
- Clinical Medicine Undergraduate Program, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Wei Zhang
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
148
|
Pappa KI, Kontostathi G, Makridakis M, Lygirou V, Zoidakis J, Daskalakis G, Anagnou NP. High Resolution Proteomic Analysis of the Cervical Cancer Cell Lines Secretome Documents Deregulation of Multiple Proteases. Cancer Genomics Proteomics 2018; 14:507-521. [PMID: 29109100 DOI: 10.21873/cgp.20060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/22/2017] [Accepted: 09/29/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Oncogenic infection by HPV, eventually leads to cervical carcinogenesis, associated by deregulation of specific pathways and protein expression at the intracellular and secretome level. Thus, secretome analysis can elucidate the biological mechanisms contributing to cervical cancer. In the present study we systematically analyzed its constitution in four cervical cell lines employing a highly sensitive proteomic technology coupled with bioinformatics analysis. MATERIALS AND METHODS LC/MS-MS proteomics and bioinformatics analysis were performed in the secretome of four informative cervical cell lines SiHa (HPV16+), HeLa (HPV18+), C33A (HPV-) and HCK1T (normal). RESULTS The proteomic pattern of each cancer cell line compared to HCK1T was identified and a detailed bioinformatics analysis disclosed inhibition of matrix metalloproteases in cancer cell lines. This prediction was further confirmed via zymography for MMP-2 and MMP-9, western blot analysis for ADAM10 and by MRM for TIMP1. The differential expression of important secreted proteins such as CATD, FUCA1 and SOD2 was also confirmed by western blot analysis. MRM-targeted proteomics analysis confirmed the differential expression of CATD, CATB, SOD2, QPCT and NEU1. CONCLUSION High resolution proteomics analysis of cervical cancer secretome revealed significantly deregulated biological processes and proteins implicated in cervical carcinogenesis.
Collapse
Affiliation(s)
- Kalliopi I Pappa
- Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece .,First Department of Obstetrics and Gynecology, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Georgia Kontostathi
- Biotechnology Division, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,Laboratory of Biology, University of Athens School of Medicine, Athens, Greece
| | - Manousos Makridakis
- Biotechnology Division, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Vasiliki Lygirou
- Biotechnology Division, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,Laboratory of Biology, University of Athens School of Medicine, Athens, Greece
| | - Jerome Zoidakis
- Biotechnology Division, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - George Daskalakis
- First Department of Obstetrics and Gynecology, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Nicholas P Anagnou
- Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,Laboratory of Biology, University of Athens School of Medicine, Athens, Greece
| |
Collapse
|
149
|
Li S, Fu L, Tian T, Deng L, Li H, Xia W, Gong Q. Disrupting SOD1 activity inhibits cell growth and enhances lipid accumulation in nasopharyngeal carcinoma. Cell Commun Signal 2018; 16:28. [PMID: 29891006 PMCID: PMC5996554 DOI: 10.1186/s12964-018-0240-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
Background SOD1 is an abundant enzyme that has been studied as a regulator of the antioxidant defence system, and this enzyme is well known for catalyzing the dismutation of superoxide into hydrogen peroxide. However the SOD1 in the progress of NPC and underlying mechanisms remain unclear. Methods In NPC tissue samples, SOD1 protein levels were measured by Western blot and immunohistochemical (IHC) staining. mRNA levels and SOD1 activity were monitored by qRT-PCR and SOD activity kit, respectively. Kaplan-Meier survival analysis was performed to explore the relationship between SOD1 expression and prognosis of NPC. The biological effects of SOD1 were investigated both in vitro by CCK-8, clonogenicity and apoptosis assays and in vivo by a xenograft mice model. Western blotting, ROS assay and triglyceride assays were applied to investigate the underlying molecular mechanism of pro-survival role of SOD1 in NPC. Results We observed a significant upregulation of SOD1 in NPC tissue and high SOD1 expression is a predictor of poor prognosis and is correlated with poor outcome. We confirmed the pro-survival role of SOD1 both in vitro and in vivo. We demonstrated that these mechanisms of SOD1 partly exist to maintain low levels of the superoxide anion and to avoid the accumulation of lipid droplets via enhanced CPT1A-mediated fatty acid oxidation. Conclusions The results of this study indicate that SOD1 is a potential prognostic biomarker and a promising target for NPC therapy. Electronic supplementary material The online version of this article (10.1186/s12964-018-0240-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuai Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Lanyan Fu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Tian Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Liwen Deng
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Huangbin Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China
| | - Weixiong Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510182, People's Republic of China.
| |
Collapse
|
150
|
Tsang CK, Chen M, Cheng X, Qi Y, Chen Y, Das I, Li X, Vallat B, Fu LW, Qian CN, Wang HY, White E, Burley SK, Zheng XFS. SOD1 Phosphorylation by mTORC1 Couples Nutrient Sensing and Redox Regulation. Mol Cell 2018; 70:502-515.e8. [PMID: 29727620 PMCID: PMC6108545 DOI: 10.1016/j.molcel.2018.03.029] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/12/2017] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
Abstract
Nutrients are not only organic compounds fueling bioenergetics and biosynthesis, but also key chemical signals controlling growth and metabolism. Nutrients enormously impact the production of reactive oxygen species (ROS), which play essential roles in normal physiology and diseases. How nutrient signaling is integrated with redox regulation is an interesting, but not fully understood, question. Herein, we report that superoxide dismutase 1 (SOD1) is a conserved component of the mechanistic target of rapamycin complex 1 (mTORC1) nutrient signaling. mTORC1 regulates SOD1 activity through reversible phosphorylation at S39 in yeast and T40 in humans in response to nutrients, which moderates ROS level and prevents oxidative DNA damage. We further show that SOD1 activation enhances cancer cell survival and tumor formation in the ischemic tumor microenvironment and protects against the chemotherapeutic agent cisplatin. Collectively, these findings identify a conserved mechanism by which eukaryotes dynamically regulate redox homeostasis in response to changing nutrient conditions.
Collapse
Affiliation(s)
- Chi Kwan Tsang
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA; Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, 613 Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Miao Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Xin Cheng
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Yanmei Qi
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Yin Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Ishani Das
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Xiaoxing Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Brinda Vallat
- Institute for Quantitative Biomedicine and Department of Chemistry and Chemical Biology, Rutgers, State University of New Jersey, Piscataway, NJ 08854 USA
| | - Li-Wu Fu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Hui-Yun Wang
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Molecular Biology and Biochemistry, Rutgers, State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Stephen K Burley
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Institute for Quantitative Biomedicine and Department of Chemistry and Chemical Biology, Rutgers, State University of New Jersey, Piscataway, NJ 08854 USA
| | - X F Steven Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA.
| |
Collapse
|