101
|
Tseng HY, Chen YA, Jen J, Shen PC, Chen LM, Lin TD, Wang YC, Hsu HL. Oncogenic MCT-1 activation promotes YY1-EGFR-MnSOD signaling and tumor progression. Oncogenesis 2017; 6:e313. [PMID: 28394354 PMCID: PMC5520490 DOI: 10.1038/oncsis.2017.13] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/10/2017] [Accepted: 02/10/2017] [Indexed: 12/20/2022] Open
Abstract
Tumor cells often produce high levels of reactive oxygen species (ROS) and display an increased ROS scavenging system. However, the molecular mechanism that balances antioxidative and oxidative stress in cancer cells is unclear. Here, we determined that oncogenic multiple copies in T-cell malignancy 1 (MCT-1) activity promotes the generation of intracellular ROS and mitochondrial superoxide. Overexpression of MCT-1 suppresses p53 accumulation but elevates the manganese-dependent superoxide dismutase (MnSOD) level via the YY1-EGFR signaling cascade, which protects cells against oxidative damage. Conversely, restricting ROS generation and/or targeting YY1 in lung cancer cells effectively inhibits the EGFR-MnSOD signaling pathway and cell invasiveness induced by MCT-1. Significantly, MCT-1 overexpression in lung cancer cells promotes tumor progression, necrosis and angiogenesis, and increases the number of tumor-promoting M2 macrophages and cancer-associated fibroblasts in the microenvironment. Clinical evidence further confirms that high expression of MCT-1 is associated with an increase in YY1, EGFR and MnSOD expression, accompanied by tumor recurrence, poor overall survival and EGFR mutation status in patients with lung cancers. Together, these data indicate that the MCT-1 oncogenic pathway is implicated in oxidative metabolism and lung carcinogenesis.
Collapse
Affiliation(s)
- H-Y Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Y-A Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - J Jen
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - P-C Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - L-M Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - T-D Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Y-C Wang
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - H-L Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| |
Collapse
|
102
|
Wang Z, Liao K, Zuo W, Liu X, Qiu Z, Gong Z, Liu C, Zeng Q, Qian Y, Jiang L, Bu Y, Hong S, Hu G. Depletion of NFBD1/MDC1 Induces Apoptosis in Nasopharyngeal Carcinoma Cells Through the p53-ROS-Mitochondrial Pathway. Oncol Res 2017; 25:123-136. [PMID: 28081741 PMCID: PMC7840771 DOI: 10.3727/096504016x14732772150226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
NFBD1, a signal amplifier of the p53 pathway, is vital for protecting cells from p53-mediated apoptosis and the early phase of DNA damage response under normal culture conditions. Here we investigated its expression in patients with nasopharyngeal carcinoma (NPC), and we describe the biological functions of the NFBD1 gene. We found that NFBD1 mRNA and protein were more highly expressed in NPC tissues than in nontumorous tissues. To investigate the function of NFBD1, we created NFBD1-depleted NPC cell lines that exhibited decreased cellular proliferation and colony formation, an increase in their rate of apoptosis, and an enhanced sensitivity to chemotherapeutic agents compared with in vitro controls. However, N-acetyl cysteine (NAC) and downregulation of p53 expression could partially reverse the apoptosis caused by the loss of NFBD1. Further analysis showed that loss of NFBD1 resulted in increased production of intracellular reactive oxygen species (ROS) depending on p53, which subsequently triggered the mitochondrial apoptotic pathway. Using a xenograft model in nude mice, we showed that silencing NFBD1 also significantly inhibited tumor growth and led to apoptosis. Taken together, our data suggest that inhibition of NFBD1 in NPC could be therapeutically useful.
Collapse
|
103
|
Abstract
The excitement around the entry into the clinic of the first generation of p53-specific drugs has become muted as the hoped-for dramatic clinical responses have not yet been seen. However, these pioneer molecules have become exceptionally powerful tools in the analysis of the p53 pathway and, as a result, a whole spectrum of new interventions are being explored. These include entirely novel and innovative approaches to drug discovery, such as the use of exon-skipping antisense oligonucleotides and T-cell-receptor-based molecules. The extraordinary resources available to the p53 community in terms of reagents, models, and collaborative networks are generating breakthrough approaches to medicines for oncology and also for other diseases in which aberrant p53 signaling plays a role.
Collapse
|
104
|
Coballase-Urrutia E, Cárdenas-Rodríguez N, González-García MC, Núñez-Ramírez E, Floriano-Sánchez E, González-Trujano ME, Fernández-Rojas B, Pedraza-Chaverrí J, Montesinos-Correa H, Rivera-Espinosa L, Sampieri AIII, Carmona-Aparicio L. Biochemical and molecular modulation of CCl 4-induced peripheral and central damage by Tilia americana var. mexicanaextracts. Saudi Pharm J 2017; 25:319-331. [PMID: 28344485 PMCID: PMC5357111 DOI: 10.1016/j.jsps.2016.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/26/2016] [Indexed: 12/13/2022] Open
Abstract
Around the world, species from the genus Tilia are commonly used because of their peripheral and central medicinal effects; they are prepared as teas and used as tranquilizing, anticonvulsant, and analgesic agents. In this study, we provide evidence of the protective effects of organic and aqueous extracts (100 mg/kg, i.p.) obtained from the leaves of Tilia americana var. mexicana on CCl4-induced liver and brain damage in the rat. Protection was observed in the liver and brain (cerebellum, cortex and cerebral hemispheres) by measuring the activity of antioxidant enzymes and levels of malondialdehyde (MDA) using spectrophotometric methods. Biochemical parameters were also assessed in serum samples from the CCl4-treated rats. The T. americana var. mexicana leaf extracts provided significant protection against CCl4-induced peripheral and central damage by increasing the activity of antioxidant enzymes, diminishing lipid peroxidation, and preventing alterations in biochemical serum parameters, such as the levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), γ-globulin (γ-GLOB), serum albumin (ALB), total bilirubin (BB), creatinine (CREA) and creatine kinase (CK), relative to the control group. Additionally, we correlated gene expression with antioxidant activity in the experimental groups treated with the organic and aqueous Tilia extracts and observed a non-statistically significant positive correlation. Our results provide evidence of the underlying biomedical properties of T. americana var. mexicana that confer its neuro- and hepatoprotective effects.
Collapse
Key Words
- ALB, serum albumin
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Ac.E, ethyl acetate extract group
- Antioxidant
- Aq.E, aqueous extract group
- Aq.E + CCl4, aqueous extract-CCl4 group
- BACT, β-actin
- BB, total bilirubin
- CAT, catalase
- CCl3OO•, trichloromethylperoxy radical
- CCl4, carbon tetrachloride
- CCl4-induced damage
- CDNB, 1-chloro-2,4-dinitrobenzene
- CK, creatine kinase
- COX-2, cyclooxygenase
- CREA, creatinine
- DMPO, 5,5-dimethyl-1-pyrrolin-N-oxide
- EDTA, ethylenediaminetetraacetic acid disodium salt
- G6PDH, glucose-6-phosphate dehydrogenase
- GAPDH, glyceraldehyde-3 phosphate dehydrogenase
- GPx, glutathione peroxidase
- GR, glutathione reductase
- GSH, reduced form of glutathione
- GSSG, oxidized form of glutathione
- GST, glutathione-S-transferase
- H2O2, hydrogen peroxide
- HO-1, heme oxygenase-1
- He.E, hexane extract group
- He.E + CCl4, hexane extract-CCl4 group
- Hepatoprotective effects
- MDA, malondialdehyde
- Me.E, methanol extract group
- Me.E + CCl4, methanol extract-CCl4 group
- NADPH, nicotinamide adenine dinucleotide phosphate
- NBT, nitro blue tetrazolium
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- Neuroprotective effects
- Nrf2, nuclear factor erythroid-derived 2-like 2
- O.O, olive oil group
- Oxidative stress
- PPARγ, peroxisome proliferator-activated receptor gamma
- RNA, ribonucleic acid
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- SOD1, superoxide dismutase-1
- SOD2, superoxide dismutase-1
- TNF-α, tumor necrosis factor
- Tilia americana var. mexicana
- UK, United Kingdom
- USA, United States of America
- Var., variant
- [Formula: see text], trichloromethyl
- bp, base pair
- i.p., intraperitoneal administration
- iNOS, inducible nitric oxide synthase
- oxo8-dG, 8-hydroxy-2′-deoxyguanosine
- γ-GLOB, γ-globulin
Collapse
Affiliation(s)
| | | | | | - Eithan Núñez-Ramírez
- Military School of Graduate of Health, Multidisciplinary Research Laboratory, SEDENA, 11270 D.F. Mexico, Mexico
| | - Esaú Floriano-Sánchez
- Military School of Graduate of Health, Multidisciplinary Research Laboratory, SEDENA, 11270 D.F. Mexico, Mexico
| | - María Eva González-Trujano
- Laboratory of Neuropharmacology of Natural Products, National Institute of Psychiatry Ramon de la Fuente Muñiz, 14370 D.F. Mexico, Mexico
| | - Berenice Fernández-Rojas
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04150 D.F. Mexico, Mexico
| | - José Pedraza-Chaverrí
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04150 D.F. Mexico, Mexico
| | | | | | - Aristides III Sampieri
- Department of Comparative Biology, Faculty of Sciences, National Autonomous University of Mexico, 04150 D.F. Mexico, Mexico
| | | |
Collapse
|
105
|
Singh MP, Han J, Kang SC. 3',5-dihydroxy-3,4',7-trimethoxyflavone-induces ER-stress-associated HCT-116 programmed cell death via redox signaling. Biomed Pharmacother 2017; 88:151-161. [PMID: 28103509 DOI: 10.1016/j.biopha.2017.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
Quercetin, a well cognized bioactive flavone possessing great medicinal value, has limited usage. The rapid gastrointestinal digestion of quercetin is also a major obstacle for its clinical implementation due to low bioavailability and poor aqueous solubility. 3',5-dihydroxy-3,4',7-trimethoxyflavone (DTMF), a novel semi-synthetic derivative of quercetin, is known to modulate several biological activities. Therefore, in the present study we examined the cytotoxic mechanism of DTMF in concentration-dependent manner (25, 50, and 100μM; 24h) against HCT-116 human colon carcinoma cells. The cytotoxic potential of DTMF was characterized based on deformed cell morphology, increased ROS accumulation, loss of mitochondrial membrane potential (ΔѰm), increased mitochondrial mass, chromatin condensation, and typical DNA-fragmentation in HCT-116 cells. The results showed that DTMF-induced enhanced ROS production at higher concentration (100μM) as evidenced by upregulated expression of ER stress and apoptotic proteins with concomitant increase in PERK, CHOP, and JNK levels, when compared to N-acetyl cysteine (NAC, ROS inhibitor) treated HCT-116 cells, which depicts that DTMF might act as a crucial mediator of apoptosis signaling. Collectively, our results suggest that DTMF stimulates ROS-mediated oxidative stress, which in turn induces PERK-CHOP and JNK pathway of apoptosis to promote HCT-116 cell death.
Collapse
Affiliation(s)
- Mahendra Pal Singh
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jaehong Han
- Metalloenzyme Research Group and Department of Integrative Plant Science, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
106
|
Tian X, Xiao BB, Wu A, Yu L, Zhou J, Wang Y, Wang N, Guan H, Shang ZF. Hydroxylated-graphene quantum dots induce cells senescence in both p53-dependent and -independent manner. Toxicol Res (Camb) 2016; 5:1639-1648. [PMID: 30090463 PMCID: PMC6061981 DOI: 10.1039/c6tx00209a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022] Open
Abstract
The numerous particular chemical/physical properties make graphene quantum dots (GQDs) attractive for various biomedical applications such as drug delivery, bioimaging and tumor photodynamic therapy (PDT). In the present study, the critical roles of hydroxyl-modified GQDs (OH-GQDs) on lung carcinoma A549 (wild type p53) and H1299 (p53-null) cells were investigated. Our data showed that a medium concentration (50 μg mL-1) of OH-GQDs significantly decreased the viability of A549 and H1299 cells. OH-GQDs treatment enhanced intracellular reactive oxygen species (ROS) generation. Furthermore, we found that treatment with ROS scavenger N-acetylcysteine (NAC) at least partially abolished the cytotoxic effect of OH-GQDs on A549 and H1299 cells. Hydroxylated GQDs lead to G0-G1 arrest and cells senescence. Signal pathway analysis revealed that OH-GQDs activated the expression of p21 in both a p53-dependent and -independent manner. Consistent with this, OH-GQDs could also inhibit the phosphorylation of Rb in both A549 and H1299 cells. These findings provide valuable information for the consideration of biomedical application of GQDs in the future.
Collapse
Affiliation(s)
- Xin Tian
- School of Radiation Medicine and Protection , Medical College of Soochow University , Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , Jiangsu 215123 , P.R. China .
| | - Bei-Bei Xiao
- School of Radiation Medicine and Protection , Medical College of Soochow University , Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , Jiangsu 215123 , P.R. China .
| | - Anqing Wu
- School of Radiation Medicine and Protection , Medical College of Soochow University , Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , Jiangsu 215123 , P.R. China .
| | - Lan Yu
- Division of Molecular Radiation Biology , Department of Radiation Oncology , University of Texas Southwestern Medical Center at Dallas , Dallas , Texas 75390 , USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory , Nanjing Medical University Affiliated Suzhou Hospital , Suzhou , Jiangsu 215001 , P.R. China
| | - Yu Wang
- Department of Radiation Toxicology and Oncology , Beijing Key Laboratory for Radiobiology (BKLRB) , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China .
| | - Nan Wang
- School of Radiation Medicine and Protection , Medical College of Soochow University , Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , Jiangsu 215123 , P.R. China .
| | - Hua Guan
- Department of Radiation Toxicology and Oncology , Beijing Key Laboratory for Radiobiology (BKLRB) , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China .
| | - Zeng-Fu Shang
- School of Radiation Medicine and Protection , Medical College of Soochow University , Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , Jiangsu 215123 , P.R. China .
| |
Collapse
|
107
|
Xing F, Zhan Q, He Y, Cui J, He S, Wang G. 1800MHz Microwave Induces p53 and p53-Mediated Caspase-3 Activation Leading to Cell Apoptosis In Vitro. PLoS One 2016; 11:e0163935. [PMID: 27689798 PMCID: PMC5045209 DOI: 10.1371/journal.pone.0163935] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 04/17/2016] [Indexed: 12/23/2022] Open
Abstract
Recent studies have reported that exposure of mammalian cells to microwave radiation may have adverse effects such as induction of cell apoptosis. However, the molecular mechanisms underlying microwave induced mammalian cell apoptosis are not fully understood. Here, we report a novel mechanism: exposure to 1800MHz microwave radiation induces p53-dependent cell apoptosis through cytochrome c-mediated caspase-3 activation pathway. We first measured intensity of microwave radiation from several electronic devices with an irradiation detector. Mouse NIH/3T3 and human U-87 MG cells were then used as receivers of 1800MHz electromagnetic radiation (EMR) at a power density of 1209 mW/m2. Following EMR exposure, cells were analyzed for viability, intracellular reactive oxygen species (ROS) generation, DNA damage, p53 expression, and caspase-3 activity. Our analysis revealed that EMR exposure significantly decreased viability of NIH/3T3 and U-87 MG cells, and increased caspase-3 activity. ROS burst was observed at 6 h and 48 h in NIH/3T3 cells, while at 3 h in U-87 MG cells. Hoechst 33258 staining and in situ TUNEL assay detected that EMR exposure increased DNA damage, which was significantly restrained in the presence of N-acetyl-L-cysteine (NAC, an antioxidant). Moreover, EMR exposure increased the levels of p53 protein and p53 target gene expression, promoted cytochrome c release from mitochondrion, and increased caspase-3 activity. These events were inhibited by pretreatment with NAC, pifithrin-α (a p53 inhibitor) and caspase inhibitor. Collectively, our findings demonstrate, for the first time, that 1800MHz EMR induces apoptosis-related events such as ROS burst and more oxidative DNA damage, which in turn promote p53-dependent caspase-3 activation through release of cytochrome c from mitochondrion. These findings thus provide new insights into physiological mechanisms underlying microwave-induced cell apoptosis.
Collapse
Affiliation(s)
- Fuqiang Xing
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Qiuqiang Zhan
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
| | - Yiduo He
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Jiesheng Cui
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Sailing He
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
- Department of Electromagnetic Engineering, Royal Institute of Technology (KTH), 10044 Stockholm, Sweden
- * E-mail: (SH); (GW)
| | - Guanyu Wang
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
- * E-mail: (SH); (GW)
| |
Collapse
|
108
|
Sanguinaria canadensis: Traditional Medicine, Phytochemical Composition, Biological Activities and Current Uses. Int J Mol Sci 2016; 17:ijms17091414. [PMID: 27618894 PMCID: PMC5037693 DOI: 10.3390/ijms17091414] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/26/2022] Open
Abstract
Sanguinaria canadensis, also known as bloodroot, is a traditional medicine used by Native Americans to treat a diverse range of clinical conditions. The plants rhizome contains several alkaloids that individually target multiple molecular processes. These bioactive compounds, mechanistically correlate with the plant’s history of ethnobotanical use. Despite their identification over 50 years ago, the alkaloids of S. canadensis have not been developed into successful therapeutic agents. Instead, they have been associated with clinical toxicities ranging from mouthwash induced leukoplakia to cancer salve necrosis and treatment failure. This review explores the historical use of S. canadensis, the molecular actions of the benzophenanthridine and protopin alkaloids it contains, and explores natural alkaloid variation as a possible rationale for the inconsistent efficacy and toxicities encountered by S.canadensis therapies. Current veterinary and medicinal uses of the plant are studied with an assessment of obstacles to the pharmaceutical development of S. canadensis alkaloid based therapeutics.
Collapse
|
109
|
Redox Homeostasis and Cellular Antioxidant Systems: Crucial Players in Cancer Growth and Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6235641. [PMID: 27418953 PMCID: PMC4932173 DOI: 10.1155/2016/6235641] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) and their products are components of cell signaling pathways and play important roles in cellular physiology and pathophysiology. Under physiological conditions, cells control ROS levels by the use of scavenging systems such as superoxide dismutases, peroxiredoxins, and glutathione that balance ROS generation and elimination. Under oxidative stress conditions, excessive ROS can damage cellular proteins, lipids, and DNA, leading to cell damage that may contribute to carcinogenesis. Several studies have shown that cancer cells display an adaptive response to oxidative stress by increasing expression of antioxidant enzymes and molecules. As a double-edged sword, ROS influence signaling pathways determining beneficial or detrimental outcomes in cancer therapy. In this review, we address the role of redox homeostasis in cancer growth and therapy and examine the current literature regarding the redox regulatory systems that become upregulated in cancer and their role in promoting tumor progression and resistance to chemotherapy.
Collapse
|
110
|
Wang X, Hai C. Novel insights into redox system and the mechanism of redox regulation. Mol Biol Rep 2016; 43:607-28. [DOI: 10.1007/s11033-016-4022-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/26/2016] [Indexed: 12/20/2022]
|
111
|
In vivo antioxidative effects of l-theanine in the presence or absence of Escherichia coli-induced oxidative stress. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
112
|
Salazar-Ramiro A, Ramírez-Ortega D, Pérez de la Cruz V, Hérnandez-Pedro NY, González-Esquivel DF, Sotelo J, Pineda B. Role of Redox Status in Development of Glioblastoma. Front Immunol 2016; 7:156. [PMID: 27199982 PMCID: PMC4844613 DOI: 10.3389/fimmu.2016.00156] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/11/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive neoplasia, prognosis remains dismal, and current therapy is mostly palliative. There are no known risk factors associated with gliomagenesis; however, it is well established that chronic inflammation in brain tissue induces oxidative stress in astrocytes and microglia. High quantities of reactive species of oxygen into the cells can react with several macromolecules, including chromosomal and mitochondrial DNA, leading to damage and malfunction of DNA repair enzymes. These changes bring genetic instability and abnormal metabolic processes, favoring oxidative environment and increase rate of cell proliferation. In GBM, a high metabolic rate and increased basal levels of reactive oxygen species play an important role as chemical mediators in the regulation of signal transduction, protecting malignant cells from apoptosis, thus creating an immunosuppressive environment. New redox therapeutics could reduce oxidative stress preventing cellular damage and high mutation rate accompanied by chromosomal instability, reducing the immunosuppressive environment. In addition, therapies directed to modulate redox rate reduce resistance and moderate the high rate of cell proliferation, favoring apoptosis of tumoral cells. This review describes the redox status in GBM, and how this imbalance could promote gliomagenesis through genomic and mitochondrial DNA damage, inducing the pro-oxidant and proinflammatory environment involved in tumor cell proliferation, resistance, and immune escape. In addition, some therapeutic agents that modulate redox status and might be advantageous in therapy against GBM are described.
Collapse
Affiliation(s)
- Aleli Salazar-Ramiro
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Daniela Ramírez-Ortega
- Neurochemistry Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | | | | | | | - Julio Sotelo
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Benjamín Pineda
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| |
Collapse
|
113
|
Sánchez-Hidalgo AC, Muñoz MF, Herrera AJ, Espinosa-Oliva AM, Stowell R, Ayala A, Machado A, Venero JL, de Pablos RM. Chronic stress alters the expression levels of longevity-related genes in the rat hippocampus. Neurochem Int 2016; 97:181-92. [PMID: 27120255 DOI: 10.1016/j.neuint.2016.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 02/02/2023]
Abstract
The molecular mechanisms underlying the negative effects of psychological stress on cellular stress during aging and neurodegenerative diseases are poorly understood. The main objective of this study was to test the effect of chronic psychological stress, and the consequent increase of circulating glucocorticoids, on several hippocampal genes involved in longevity. Sirtuin-1, p53, thioredoxin-interacting protein, and heat shock protein 70 were studied at the mRNA and protein levels in stressed and non-stressed animals. Stress treatment for 10 days decreased sirtuin-1 and heat shock protein 70 levels, but increased levels of p53, thioredoxin-interacting protein and the NADPH oxidase enzyme. Examination of protein expression following two months of stress treatment indicated that sirtuin-1 remained depressed. In contrast, an increase was observed for thioredoxin-interacting protein, heat shock protein 70, p53 and the NADPH oxidase enzyme. The effect of stress was reversed by mifepristone, a glucocorticoid receptor antagonist. These data suggest that chronic stress could contribute to aging in the hippocampus.
Collapse
Affiliation(s)
- Ana C Sánchez-Hidalgo
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Mario F Muñoz
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Antonio J Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Ana M Espinosa-Oliva
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Rianne Stowell
- Department of Neuroscience, University of Rochester Medical Center, 601 Elmwood Avenue, Box 603, Rochester, NY 14642, USA
| | - Antonio Ayala
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Alberto Machado
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - José L Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Rocío M de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain.
| |
Collapse
|
114
|
Jin H, Yin S, Song X, Zhang E, Fan L, Hu H. p53 activation contributes to patulin-induced nephrotoxicity via modulation of reactive oxygen species generation. Sci Rep 2016; 6:24455. [PMID: 27071452 PMCID: PMC4829895 DOI: 10.1038/srep24455] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/29/2016] [Indexed: 11/09/2022] Open
Abstract
Patulin is a major mycotoxin found in fungal contaminated fruits and their derivative products. Previous studies showed that patulin was able to induce increase of reactive oxygen species (ROS) generation and oxidative stress was suggested to play a pivotal role in patulin-induced multiple toxic signaling. The objective of the present study was to investigate the functional role of p53 in patulin-induced oxidative stress. Our study demonstrated that higher levels of ROS generation and DNA damage were induced in wild-type p53 cell lines than that found in either knockdown or knockout p53 cell lines in response to patulin exposure, suggesting p53 activation contributed to patulin-induced ROS generation. Mechanistically, we revealed that the pro-oxidant role of p53 in response to patulin was attributed to its ability to suppress catalase activity through up-regulation of PIG3. Moreover, these in vitro findings were further validated in the p53 wild-type/knockout mouse model. To the best of our knowledge, this is the first report addressing the functional role of p53 in patulin-induced oxidative stress. The findings of the present study provided novel insights into understanding mechanisms behind oxidative stress in response to patulin exposure.
Collapse
Affiliation(s)
- Huan Jin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, No17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Shutao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, No17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Xinhua Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, No17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Enxiang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, No17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Lihong Fan
- College of Veterinary Medicine, China Agricultural University, No2 Yunamingyuan West Road, Haidian District, Beijing 100193, China
| | - Hongbo Hu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, No17 Qinghua East Road, Haidian District, Beijing 100083, China
| |
Collapse
|
115
|
Arnold MC, Forte JE, Osterberg JS, Di Giulio RT. Antioxidant Rescue of Selenomethionine-Induced Teratogenesis in Zebrafish Embryos. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2016; 70:311-20. [PMID: 26498942 PMCID: PMC4842345 DOI: 10.1007/s00244-015-0235-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 10/07/2015] [Indexed: 05/23/2023]
Abstract
Selenium (Se) is an essential micronutrient that can be found at toxic concentrations in surface waters contaminated by runoff from agriculture and coal mining. Zebrafish (Danio rerio) embryos were exposed to aqueous Se in the form of selenate, selenite, and l-selenomethionine (SeMet) in an attempt to determine if oxidative stress plays a role in selenium embryo toxicity. Selenate and selenite exposure did not induce embryo deformities (lordosis and craniofacial malformation). l-selenomethionine, however, induced significantly higher deformity rates at 100 µg/L compared with controls. SeMet exposure induced a dose-dependent increase in the catalytic subunit of glutamate-cysteine ligase (gclc) and reached an 11.7-fold increase at 100 µg/L. SeMet exposure also reduced concentrations of TGSH, RGSH, and the TGSH:GSSG ratio. Pretreatment with 100 µM N-acetylcysteine significantly reduced deformities in the zebrafish embryos secondarily treated with 400 µg/L SeMet from approximately 50–10 % as well as rescued all three of the significant glutathione level differences seen with SeMet alone. Selenite exposure induced a 6.6-fold increase in expression of the glutathione-S-transferase pi class 2 (gstp2) gene, which is involved in xenobiotic transformation and possibly oxidative stress. These results suggest that aqueous exposure to SeMet can induce significant embryonic teratogenesis in zebrafish that are at least partially attributed to oxidative stress.
Collapse
Affiliation(s)
- M C Arnold
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC 27708, USA.
| | | | | | | |
Collapse
|
116
|
Brault C, Lévy P, Duponchel S, Michelet M, Sallé A, Pécheur EI, Plissonnier ML, Parent R, Véricel E, Ivanov AV, Demir M, Steffen HM, Odenthal M, Zoulim F, Bartosch B. Glutathione peroxidase 4 is reversibly induced by HCV to control lipid peroxidation and to increase virion infectivity. Gut 2016; 65:144-154. [PMID: 25516417 DOI: 10.1136/gutjnl-2014-307904] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/19/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Inflammation and oxidative stress drive disease progression in chronic hepatitis C (CHC) towards hepatocellular carcinoma. HCV is known to increase intracellular levels of reactive oxygen species (ROS), but how it eliminates ROS is less well known. The role of the ROS scavenger glutathione peroxidase 4 (GPx4), induced by HCV, in the viral life cycle was analysed. DESIGN The study was performed using a replicative in vitro HCV infection model and liver biopsies derived from two different CHC patient cohorts. RESULTS A screen for HCV-induced peroxide scavengers identified GPx4 as a host factor required for HCV infection. The physiological role of GPx4 is the elimination of lipid peroxides from membranes or lipoproteins. GPx4-silencing reduced the specific infectivity of HCV by up to 10-fold. Loss of infectivity correlated with 70% reduced fusogenic activity of virions in liposome fusion assays. NS5A was identified as the protein that mediates GPx4 induction in a phosphatidylinositol-3-kinase-dependent manner. Levels of GPx4 mRNA were found increased in vitro and in CHC compared with control liver biopsies. Upon successful viral eradication, GPx4 transcript levels returned to baseline in vitro and also in the liver of patients. CONCLUSIONS HCV induces oxidative stress but controls it tightly by inducing ROS scavengers. Among these, GPx4 plays an essential role in the HCV life cycle. Modulating oxidative stress in CHC by specifically targeting GPx4 may lower specific infectivity of virions and prevent hepatocarcinogenesis, especially in patients who remain difficult to be treated in the new era of interferon-free regimens.
Collapse
Affiliation(s)
- Charlène Brault
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | - Pierre Lévy
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | - Sarah Duponchel
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | - Maud Michelet
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | - Aurèlie Sallé
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | | | | | - Romain Parent
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France DevWeCan Laboratories of Excellence Network (Labex), France
| | - Evelyne Véricel
- Université de Lyon, UMR 1060 INSERM CarMeN, IMBL, INSA-Lyon, Lyon, France
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Münevver Demir
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | | | - Fabien Zoulim
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France DevWeCan Laboratories of Excellence Network (Labex), France Hospices civils de Lyon, Lyon, France
| | - Birke Bartosch
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France DevWeCan Laboratories of Excellence Network (Labex), France
| |
Collapse
|
117
|
Wang X, Hai C. Redox modulation of adipocyte differentiation: hypothesis of "Redox Chain" and novel insights into intervention of adipogenesis and obesity. Free Radic Biol Med 2015; 89:99-125. [PMID: 26187871 DOI: 10.1016/j.freeradbiomed.2015.07.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/19/2015] [Accepted: 07/08/2015] [Indexed: 02/08/2023]
Abstract
In view of the global prevalence of obesity and obesity-associated disorders, it is important to clearly understand how adipose tissue forms. Accumulating data from various laboratories implicate that redox status is closely associated with energy metabolism. Thus, biochemical regulation of the redox system may be an attractive alternative for the treatment of obesity-related disorders. In this work, we will review the current data detailing the role of the redox system in adipocyte differentiation, as well as identifying areas for further research. The redox system affects adipogenic differentiation in an extensive way. We propose that there is a complex and interactive "redox chain," consisting of a "ROS-generating enzyme chain," "combined antioxidant chain," and "transcription factor chain," which contributes to fine-tune the regulation of ROS level and subsequent biological consequences. The roles of the redox system in adipocyte differentiation are paradoxical. The redox system exerts a "tridimensional" mechanism in the regulation of adipocyte differentiation, including transcriptional, epigenetic, and posttranslational modulations. We suggest that redoxomic techniques should be extensively applied to understand the biological effects of redox alterations in a more integrated way. A stable and standardized "redox index" is urgently needed for the evaluation of the general redox status. Therefore, more effort should be made to establish and maintain a general redox balance rather than to conduct simple prooxidant or antioxidant interventions, which have comprehensive implications.
Collapse
Affiliation(s)
- Xin Wang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
118
|
Paul S, Jakhar R, Bhardwaj M, Kang SC. Glutathione-S-transferase omega 1 (GSTO1-1) acts as mediator of signaling pathways involved in aflatoxin B1-induced apoptosis-autophagy crosstalk in macrophages. Free Radic Biol Med 2015; 89:1218-30. [PMID: 26561775 DOI: 10.1016/j.freeradbiomed.2015.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/19/2015] [Accepted: 11/04/2015] [Indexed: 12/29/2022]
Abstract
Aflatoxin B1 (AFB1) is the most toxic aflatoxin species and has been shown to be associated with specific as well as non-specific immune responses. In the present study, using murine macrophage Raw 264.7 cells as a model, we report that short exposure (6h) to AFB1 caused an increase in the cellular calcium pool in mitochondria, which in turn elevated reactive oxygen species (ROS)-mediated oxidative stress and led to loss of mitochondrial membrane potential and ultimately c-Jun N-terminal kinases (JNK)-mediated caspase-dependent cell death. On the contrary, longer exposure (12h) to AFB1 reduced JNK phosphorylation and cell death in macrophages. Measurement of autophagic flux demonstrated that autophagy induction through the canonical pathway was responsible for suppressing AFB1-induced apoptosis after 12h. As a detailed molecular mechanism, we found that the unfolded protein response (UPR) machinery was active at 12h post-exposure to AFB1 and induced cytoprotective autophagy as confirmed by determination of major autophagic markers. Inhibition of autophagy by Beclin-1 siRNA also resulted in JNK-mediated cell death. We further established that glutathione S transferase omega1-1 (GSTO1-1), a specific class of GST, was the responsible factor between apoptosis and autophagy crosstalk. Targeting of GSTO1-1 increased JNK-mediated apoptosis by 2-fold compared to the control, whereas autophagy rate was reduced. Thus, increased expression of GSTO1-1 was associated with increased protein glutathionylation, an important protein modification in response to cellular redox status.
Collapse
Affiliation(s)
- Souren Paul
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Rekha Jakhar
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Monika Bhardwaj
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea.
| |
Collapse
|
119
|
Memmert S, Gölz L, Pütz P, Jäger A, Deschner J, Appel T, Baumgarten G, Rath-Deschner B, Frede S, Götz W. Regulation of p53 under hypoxic and inflammatory conditions in periodontium. Clin Oral Investig 2015; 20:1781-9. [PMID: 26620730 DOI: 10.1007/s00784-015-1679-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/22/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Different studies suggest that inflammation as well as hypoxia leads to an increase of p53 protein levels. However, the implication of p53 during oral inflammatory processes is still unknown. The aim of this study was therefore to investigate the effect of hypoxia and inflammation on p53 regulation in human periodontium in vitro and in vivo. MATERIALS AND METHODS Under hypoxic and normoxic conditions, human primary periodontal ligament (PDL) fibroblasts (n = 9) were stimulated with lipopolysaccharides (LPS) from Porphyromonas gingivalis (P.g.), a periodontal pathogenic bacterium. After different time points, cell viability was tested; p53 gene expression, protein synthesis, and activation were measured using quantitative RT-PCR, immunoblotting, and immunofluorescence. Moreover, healthy and inflamed periodontal tissues were obtained from 12 donors to analyze p53 protein in oral inflammatory diseases by immunohistochemistry. RESULTS LPS-P.g. and hypoxia initially induced a significant upregulation of p53 mRNA expression and p53 protein levels. Nuclear translocation of p53 after inflammatory stimulation supported these findings. Hypoxia first enhanced p53 levels, but after 24 h of incubation, protein levels decreased, which was accompanied by an improvement of PDL cell viability. Immunohistochemistry revealed an elevation of p53 immunoreactivity in accordance to the progression of periodontal inflammation. CONCLUSIONS Our data indicate that p53 plays a pivotal role in PDL cell homeostasis and seems to be upregulated in oral inflammatory diseases. CLINICAL RELEVANCE Upregulation of p53 may promote the destruction of periodontal integrity. A possible relationship with carcinogenesis may be discussed.
Collapse
Affiliation(s)
- S Memmert
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany. .,Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Bonn, Germany.
| | - L Gölz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - P Pütz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - A Jäger
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - J Deschner
- Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - T Appel
- Clinic of Oral and Maxillofacial Surgery, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - G Baumgarten
- Clinic and Polyclinic of Anesthesiology and Intensive Care Medicine, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - B Rath-Deschner
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - S Frede
- Clinic and Polyclinic of Anesthesiology and Intensive Care Medicine, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - W Götz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, Faculty of Medicine, University of Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| |
Collapse
|
120
|
AlGhamdi S, Leoncikas V, Plant KE, Plant NJ. Synergistic interaction between lipid-loading and doxorubicin exposure in Huh7 hepatoma cells results in enhanced cytotoxicity and cellular oxidative stress: implications for acute and chronic care of obese cancer patients. Toxicol Res (Camb) 2015; 4:1479-1487. [PMID: 26744621 PMCID: PMC4692330 DOI: 10.1039/c5tx00173k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/11/2015] [Indexed: 01/29/2023] Open
Abstract
There has been a dramatic increase in the number of clinically obese individuals in the last twenty years. This has resulted in an increasingly common scenario where obese individuals are treated for other diseases, including cancer. Here, we examine interactions between lipid-induced steatosis and doxorubicin treatment in the human hepatoma cell line Huh7. The response of cells to either doxorubicin, lipid-loading or a combination were examined at the global level by DNA microarray, and for specific endpoints of cytotoxicity, lipid-loading, reactive oxygen species, anti-oxidant response systems, and apoptosis. Both doxorubicin and lipid-loading caused a significant accumulation of lipid within Huh7 cells, with the combination resulting in an additive accumulation. In contrast, cytotoxicity was synergistic for the combination compared to the individual components, suggesting an enhanced sensitivity of lipid-loaded cells to the acute hepatotoxic effects of doxorubicin. We demonstrate that a synergistic increase in reactive oxygen species and deregulation of protective anti-oxidant systems, most notably metallothionein expression, underlies this effect. Transcriptome analysis confirms synergistic changes at the global level, and is consistent with enhanced pro-inflammatory signalling in steatotic cells challenged with doxorubicin. Such effects are consistent with a potentiation of progression along the fatty liver disease spectrum. This suggests that treatment of obese individuals with doxorubicin may increase the risk of both acute (i.e. hepatotoxicity) and chronic (i.e. progress of fatty liver disease) adverse effects. This work highlights the need for more study in the growing therapeutic area to develop risk mitigation strategies.
Collapse
Affiliation(s)
- S AlGhamdi
- Department of Biochemistry and Physiology , Faculty of Health and Medical Sciences , University of Surrey , Guildford , Surrey GU2 7XH , UK .
| | - V Leoncikas
- Department of Biochemistry and Physiology , Faculty of Health and Medical Sciences , University of Surrey , Guildford , Surrey GU2 7XH , UK .
| | - K E Plant
- Department of Biochemistry and Physiology , Faculty of Health and Medical Sciences , University of Surrey , Guildford , Surrey GU2 7XH , UK .
| | - N J Plant
- Department of Biochemistry and Physiology , Faculty of Health and Medical Sciences , University of Surrey , Guildford , Surrey GU2 7XH , UK .
| |
Collapse
|
121
|
Manganese superoxide dismutase (MnSOD) is a malignant astrocytoma specific biomarker and associated with adverse prognosis in p53 expressing glioblastoma. Pathol Res Pract 2015; 212:17-23. [PMID: 26616112 DOI: 10.1016/j.prp.2015.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 10/27/2015] [Accepted: 11/03/2015] [Indexed: 01/24/2023]
Abstract
BACKGROUND Manganese super oxide dismutase (MnSOD) has been previously identified as one of the top regulated genes associated with poor survival in glioblastoma (GBM) patients. In the current study we have evaluated the protein expression of MnSOD across various grades of astrocytoma, studied its influence on survival of GBM patients and following recurrence. METHODS The protein expression of MnSOD was analyzed on tumor tissue sections by immunohistochemistry on 30 diffuse astrocytomas (DA), 50 anaplastic astrocytomas (AA), 30 paired (primary and recurrent) GBM samples and 30 non-tumor brain tissues. The protein expression among the different grades of diffusely infiltrating astrocytoma (DIA) was evaluated by Kruskal-Wallis one-way ANOVA followed by post hoc test. Wilcoxon matched pair test was employed to assess MnSOD protein expression across 30 paired GBM samples (primary and recurrent). The prognostic impact of MnSOD protein expression individually and following stratification with p53 expression was evaluated in a cohort of 123 GBM patients. Both over-all survival (OS) and progression free survival (PFS) analysis were performed by employing Cox regression analysis and Kaplan-Meier survival analysis on GBM patients. RESULTS A significantly increased protein expression of MnSOD was observed among malignant astrocytomas (GBM and AA) in comparison with either DA or non-tumor brain tissues (p<0.05). Among the GBM cases it was noted that the IDH1 immunopositive tumors (R132H mutant protein; n=17) had a low MnSOD expression as opposed to IDH1 immunonegative tumors (n=106), which had high expression of MnSOD (p=0.0307). Further, a statistically significant increase (p=0.010) in extent of MnSOD protein expression was also noted in GBM tumors following recurrence. Protein expression of MnSOD was associated with both poor OS (HR: 1.021; p=0.011) and early PFS (HR: 1.022; p=0.006) on univariate analysis. Multivariate Cox regression analysis as well as Kaplan-Meier survival analysis demonstrated similar poor prognostic association. Stratification of GBM cases based on p53 expression status revealed a strong association of MnSOD with OS (HR: 1.042; p=0.002) and PFS (HR: 1.044; p=0.001) in p53 positive tumor tissue samples. Similar findings were noted on multivariate Cox regression analysis and K-M survival analysis, while no such association was noted in tumor tissues staining negative for p53 expression. CONCLUSIONS Our study shows an increased expression of MnSOD in anaplastic astrocytoma and GBM compared to low grade astrocytoma and control brain. An increase in MnSOD expression following GBM tumor recurrence strengthens its putative role in tumor aggressiveness. Further, MnSOD emerges as a poor prognostic biomarker in p53 expressing GBMs, rendering this molecule as a potential therapeutic target in such patients.
Collapse
|
122
|
Dual regulation of energy metabolism by p53 in human cervix and breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3266-78. [PMID: 26434996 DOI: 10.1016/j.bbamcr.2015.09.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/25/2015] [Accepted: 09/30/2015] [Indexed: 12/14/2022]
Abstract
The role of p53 as modulator of OxPhos and glycolysis was analyzed in HeLa-L (cells containing negligible p53 protein levels) and HeLa-H (p53-overexpressing) human cervix cancer cells under normoxia and hypoxia. In normoxia, functional p53, mitochondrial enzyme contents, mitochondrial electrical potential (ΔΨm) and OxPhos flux increased in HeLa-H vs. HeLa-L cells; whereas their glycolytic enzyme contents and glycolysis flux were unchanged. OxPhos provided more than 70% of the cellular ATP and proliferation was abolished by anti-mitochondrial drugs in HeLa-H cells. In hypoxia, both cell proliferations were suppressed, but HeLa-H cells exhibited a significant decrease in OxPhos protein contents, ΔΨm and OxPhos flux. Although glycolytic function was also diminished vs. HeLa-L cells in hypoxia, glycolysis provided more than 60% of cellular ATP in HeLa-H cells. The energy metabolism phenotype of HeLa-H cells was reverted to that of HeLa-L cells by incubating with pifithrin-α, a p53-inhibitor. In normoxia, the energy metabolism phenotype of breast cancer MCF-7 cells was similar to that of HeLa-H cells, whereas p53shRNAMCF-7 cells resembled the HeLa-L cell phenotype. In hypoxia, autophagy proteins and lysosomes contents increased 2-5 times in HeLa-H cells suggesting mitophagy activation. These results indicated that under normoxia p53 up-regulated OxPhos without affecting glycolysis, whereas under hypoxia, p53 down-regulated both OxPhos (severely) and glycolysis (weakly). These p53 effects appeared mediated by the formation of p53-HIF-1α complexes. Therefore, p53 exerts a dual and contrasting regulatory role on cancer energy metabolism, depending on the O₂level.
Collapse
|
123
|
High temperature induces apoptosis and oxidative stress in pufferfish (Takifugu obscurus) blood cells. J Therm Biol 2015; 53:172-9. [DOI: 10.1016/j.jtherbio.2015.08.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/25/2015] [Accepted: 08/03/2015] [Indexed: 01/20/2023]
|
124
|
Park CM, Kim MJ, Kim SM, Park JH, Kim ZH, Choi YS. Umbilical cord mesenchymal stem cell-conditioned media prevent muscle atrophy by suppressing muscle atrophy-related proteins and ROS generation. In Vitro Cell Dev Biol Anim 2015; 52:68-76. [DOI: 10.1007/s11626-015-9948-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/31/2015] [Indexed: 12/11/2022]
|
125
|
Daglar K, Biberoglu E, Kirbas A, Dirican AO, Genc M, Avci A, Biberoglu K. The cellular immunity and oxidative stress markers in early pregnancy loss. J Matern Fetal Neonatal Med 2015; 29:1840-3. [PMID: 26135767 DOI: 10.3109/14767058.2015.1064886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE We investigated whether changes in cellular immunity and oxidative stress in pregnancy have any association with spontaneous miscarriage. MATERIAL AND METHODS Circulating adenosine deaminase (ADA) activity as a marker of cellular immunity and malondialdehyde (MDA) and catalase (CAT), glutathione peroxidase (GPx) as markers of T lymphocyte activation and parameters of oxidative stress and antioxidant defense were compared between 40 women with early pregnancy loss and another 40 women with ungoing healthy pregnancy. RESULTS Women with miscarriage had higher serum ADA and GPx levels when compared with women with normal pregnancy (p = 0.034 and p < 0.001, respectively). Although serum MDA level was slightly higher in women with miscarriage, the difference was not significant (p = 0.083). CAT levels were alike in both groups. CONCLUSION We have demonstrated an increased cellular immunity and perhaps a compensated oxidative stress related to increased antioxidant activation in women with early spontaneous pregnancy loss.
Collapse
Affiliation(s)
- Korkut Daglar
- a Department of Perinatology , Zekai Tahir Burak Women Health Care, Education and Research Hospital , Ankara , Turkey
| | - Ebru Biberoglu
- a Department of Perinatology , Zekai Tahir Burak Women Health Care, Education and Research Hospital , Ankara , Turkey
| | - Ayse Kirbas
- a Department of Perinatology , Zekai Tahir Burak Women Health Care, Education and Research Hospital , Ankara , Turkey
| | - Aylin Onder Dirican
- a Department of Perinatology , Zekai Tahir Burak Women Health Care, Education and Research Hospital , Ankara , Turkey
| | - Metin Genc
- b Department of Biochemistry , Ankara University Faculty of Medicine , Ankara , Turkey , and
| | - Aslihan Avci
- b Department of Biochemistry , Ankara University Faculty of Medicine , Ankara , Turkey , and
| | - Kutay Biberoglu
- c Department of Obstetrics and Gynecology , Gazi University Medical School , Ankara , Turkey
| |
Collapse
|
126
|
Braga EA, Khodyrev DS, Loginov VI, Pronina IV, Senchenko VN, Dmitriev AA, Kubatiev AA, Kushlinskii NE. Methylation in the regulation of the expression of chromosome 3 and microRNA genes in clear-cell renal cell carcinomas. RUSS J GENET+ 2015. [DOI: 10.1134/s1022795415050026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
127
|
Quercetin and Tryptanthrin: Two Broad Spectrum Anticancer Agents for Future Chemotherapeutic Interventions. Enzymes 2015; 37:43-72. [PMID: 26298455 DOI: 10.1016/bs.enz.2015.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The idea and practice of developing or identifying compounds capable of eliminating the transformed cells or cancer cells without being nontoxic to their normal counterparts deserves much importance. Since ages, plants have been considered and proven to be repertoires of chemicals possessing immense therapeutic potential. A proportion of these plant-derived compounds or phytochemicals were shown to be highly competent anticancer agents besides being effective against many other diseases. Representative compounds of different classes of phytochemicals are in clinical use against cancer. In this chapter, we discuss the anticancer potential of two compounds: quercetin, a flavonoid and tryptanthrin, an indoloquinazoline alkaloid, and the mechanisms behind their cytotoxic effects on cancers of different origin. The chapter also gives a brief mention of their properties that make them effective against cancer.
Collapse
|
128
|
Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Biol 2015; 16:393-405. [PMID: 26122615 DOI: 10.1038/nrm4007] [Citation(s) in RCA: 805] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The function of p53 as a tumour suppressor has been attributed to its ability to promote cell death or permanently inhibit cell proliferation. However, in recent years, it has become clear that p53 can also contribute to cell survival. p53 regulates various metabolic pathways, helping to balance glycolysis and oxidative phosphorylation, limiting the production of reactive oxygen species, and contributing to the ability of cells to adapt to and survive mild metabolic stresses. Although these activities may be integrated into the tumour suppressive functions of p53, deregulation of some elements of the p53-induced response might also provide tumours with a survival advantage.
Collapse
Affiliation(s)
- Flore Kruiswijk
- 1] Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK. [2]
| | | | - Karen H Vousden
- Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
129
|
Jeong SG, Cho GW. Endogenous ROS levels are increased in replicative senescence in human bone marrow mesenchymal stromal cells. Biochem Biophys Res Commun 2015; 460:971-6. [DOI: 10.1016/j.bbrc.2015.03.136] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 01/16/2023]
|
130
|
p53-mediated autophagic regulation: A prospective strategy for cancer therapy. Cancer Lett 2015; 363:101-7. [PMID: 25896632 DOI: 10.1016/j.canlet.2015.04.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 12/25/2022]
Abstract
Autophagy is a major catabolic process that degrades and recycles cytosolic components in autophagosomes, which fuse with lysosomes. This process enables starving cells to sustain their energy requirements and metabolic states, thus facilitating their survival, especially in cancer pathogenesis. The regulation of autophagy is quite intricate. It involves a series of signaling cascades including p53, known as the best-characterized tumor suppressor protein. Recent reports have indicated that p53 plays dual roles in regulating autophagy depending on its subcellular localization. Nuclear p53 facilitates autophagy by transactivating its target genes, whereas cytoplasmic p53 mainly inhibits autophagy through extranuclear, transcription-independent mechanisms. The relationship between autophagy and neoplasia is complicated. It may be intrinsically associated with the functional status of p53, but this is not clearly elucidated. This review focuses on the role of p53 as a master regulator of autophagy. We conclude that the contextual role of autophagy in cancer, which could be switched by p53 status, is expected to be developed into a new anticancer therapeutic approach.
Collapse
|
131
|
Witalison EE, Cui X, Hofseth AB, Subramanian V, Causey CP, Thompson PR, Hofseth LJ. Inhibiting protein arginine deiminases has antioxidant consequences. J Pharmacol Exp Ther 2015; 353:64-70. [PMID: 25635139 PMCID: PMC4366755 DOI: 10.1124/jpet.115.222745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/28/2015] [Indexed: 01/16/2023] Open
Abstract
Ulcerative colitis is a dynamic, idiopathic, chronic inflammatory condition that carries a high colon cancer risk. We previously showed that Cl-amidine, a small-molecule inhibitor of the protein arginine deiminases, suppresses colitis in mice. Because colitis is defined as inflammation of the colon associated with infiltration of white blood cells that release free radicals and citrullination is an inflammation-dependent process, we asked whether Cl-amidine has antioxidant properties. Here we show that colitis induced with azoxymethane via intraperitoneal injection + 2% dextran sulfate sodium in the drinking water is suppressed by Cl-amidine (also given in the drinking water). Inducible nitric oxide synthase, an inflammatory marker, was also downregulated in macrophages by Cl-amidine. Because epithelial cell DNA damage associated with colitis is at least in part a result of an oxidative burst from overactive leukocytes, we tested the hypothesis that Cl-amidine can inhibit leukocyte activation, as well as subsequent target epithelial cell DNA damage in vitro and in vivo. Results are consistent with this hypothesis, and because DNA damage is a procancerous mechanism, our data predict that Cl-amidine will not only suppress colitis, but we hypothesize that it may prevent colon cancer associated with colitis.
Collapse
Affiliation(s)
- Erin E Witalison
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Xiangli Cui
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Anne B Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Venkataraman Subramanian
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Corey P Causey
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Paul R Thompson
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina (E.E.W., X.C., A.B.H., L.J.H.); Shanxi Medical University, Taiyuan, Shanxi, China (X.C.); Department of Chemistry, The Scripps Research Institute, Scripps Florida, Jupiter, Florida (V.S.); Department of Chemistry, University of North Florida, Jacksonville, Florida (C.P.C.); and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (P.R.T.)
| |
Collapse
|
132
|
Moretti D, Del Bello B, Allavena G, Corti A, Signorini C, Maellaro E. Calpain-3 impairs cell proliferation and stimulates oxidative stress-mediated cell death in melanoma cells. PLoS One 2015; 10:e0117258. [PMID: 25658320 PMCID: PMC4319969 DOI: 10.1371/journal.pone.0117258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/19/2014] [Indexed: 11/18/2022] Open
Abstract
Calpain-3 is an intracellular cysteine protease, belonging to Calpain superfamily and predominantly expressed in skeletal muscle. In human melanoma cell lines and biopsies, we previously identified two novel splicing variants (hMp78 and hMp84) of Calpain-3 gene (CAPN3), which have a significant lower expression in vertical growth phase melanomas and, even lower, in metastases, compared to benign nevi. In the present study, in order to investigate the pathophysiological role played by the longer Calpain-3 variant, hMp84, in melanoma cells, we over-expressed it in A375 and HT-144 cells. In A375 cells, the enforced expression of hMp84 induces p53 stabilization, and modulates the expression of a few p53- and oxidative stress-related genes. Consistently, hMp84 increases the intracellular production of ROS (Reactive Oxygen Species), which lead to oxidative modification of phospholipids (formation of F2-isoprostanes) and DNA damage. Such events culminate in an adverse cell fate, as indicated by the decrease of cell proliferation and by cell death. To a different extent, either the antioxidant N-acetyl-cysteine or the p53 inhibitor, Pifithrin-α, recover cell viability and decrease ROS formation. Similarly to A375 cells, hMp84 over-expression causes inhibition of cell proliferation, cell death, and increase of both ROS levels and F2-isoprostanes also in HT-144 cells. However, in these cells no p53 accumulation occurs. In both cell lines, no significant change of cell proliferation and cell damage is observed in cells over-expressing the mutant hMp84C42S devoid of its enzymatic activity, suggesting that the catalytic activity of hMp84 is required for its detrimental effects. Since a more aggressive phenotype is expected to benefit from down-regulation of mechanisms impairing cell growth and survival, we envisage that Calpain-3 down-regulation can be regarded as a novel mechanism contributing to melanoma progression.
Collapse
Affiliation(s)
- Daniele Moretti
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Istituto Toscano Tumori (ITT), Firenze, Italy
| | - Barbara Del Bello
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Istituto Toscano Tumori (ITT), Firenze, Italy
| | - Giulia Allavena
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Istituto Toscano Tumori (ITT), Firenze, Italy
| | - Alessandro Corti
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Pisa, Italy
- Istituto Toscano Tumori (ITT), Firenze, Italy
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emilia Maellaro
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Istituto Toscano Tumori (ITT), Firenze, Italy
- * E-mail:
| |
Collapse
|
133
|
Chan ASL, Mowla SN, Arora P, Jat PS. Tumour suppressors and cellular senescence. IUBMB Life 2014; 66:812-22. [PMID: 25557529 DOI: 10.1002/iub.1335] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/01/2014] [Indexed: 01/09/2023]
Abstract
Cellular senescence is a stable cell cycle arrest that normal cells undergo in response to a variety of intrinsic and extrinsic stimuli, including progressive telomere shortening, changes in telomeric structure or other forms of genotoxic as well nongenotoxic stress. Senescence is thought to have originated as a remodelling program that is active in embryonic development and acts as a key tumour suppressor mechanism during the reproductive stage in early adult life, by leading to the removal of potentially cancerous cells. However, in later adult life, it promotes organismal aging by compromising tissue repair and regeneration due to the accumulation of senescent cells, depletion of stem/progenitor cells and secretion of an array of inflammatory cytokines, chemokines and matrix metalloproteases. Whilst suppressing tumour formation in the senescent cells, these inflammatory cytokines, chemokines and metalloproteases can promote tumour progression and metastasis in the neighbouring cells. Herein, we review the molecular pathways that underlie cellular senescence and how it contributes towards tumour suppression.
Collapse
Affiliation(s)
- Adelyne S L Chan
- Department of Neurodegenerative Disease and MRC Prion Unit, UCL Institute of Neurology, Queen Square, London, WC1N 3BG
| | | | | | | |
Collapse
|
134
|
Budanov AV. The role of tumor suppressor p53 in the antioxidant defense and metabolism. Subcell Biochem 2014; 85:337-58. [PMID: 25201203 DOI: 10.1007/978-94-017-9211-0_18] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor suppressor p53 is inactivated in most cancers and the critical role of p53 in the suppression of carcinogenesis has been confirmed in many mouse models. The protein product of the tumor suppressor p53 gene works as a transcriptional regulator, activating expression of numerous genes involved in cell death, cell cycle arrest, senescence, DNA-repair and many other processes. In spite of the multiple efforts to characterize the functions of p53, the mechanisms of tumor suppression by p53 are still elusive. Recently, new activities of p53 such as regulation of reactive oxygen species (ROS) and metabolism have been described and the p53-regulated genes responsible for these functions have been identified. Metabolic derangements and accumulation of ROS are features of carcinogenesis, supporting the idea that many tumor suppressive effects of p53 can be mediated by regulation of metabolism and/or ROS. Mutations in the p53 gene can not only inactivate wild type function of p53 but also endow p53 with new functions such as activation of new metabolic pathways contributing to carcinogenesis. Understanding the metabolic and antioxidant functions of p53 allows us to develop approaches to restore p53 function in cancers, where p53 is inactivated, in other to ensure the best outcome of anti-cancer treatment.
Collapse
Affiliation(s)
- Andrei V Budanov
- Department of Neurosurgery & Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA,
| |
Collapse
|
135
|
Becuwe P, Ennen M, Klotz R, Barbieux C, Grandemange S. Manganese superoxide dismutase in breast cancer: from molecular mechanisms of gene regulation to biological and clinical significance. Free Radic Biol Med 2014; 77:139-51. [PMID: 25224035 DOI: 10.1016/j.freeradbiomed.2014.08.026] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 01/06/2023]
Abstract
Breast cancer is one of the most common malignancies of all cancers in women worldwide. Many difficulties reside in the prediction of tumor metastatic progression because of the lack of sufficiently reliable predictive biological markers, and this is a permanent preoccupation for clinicians. Manganese superoxide dismutase (MnSOD) may represent a rational candidate as a predictive biomarker of breast tumor metastatic progression, because its gene expression is profoundly altered between early and advanced breast cancer, in contrast to expression in the normal mammary gland. In this review, we report the characterization of some gene polymorphisms and molecular mechanisms of SOD2 gene regulation, which allows a better understanding of how MnSOD is decreased in early breast cancer and increased in advanced breast cancer. Several studies display the biological significance of MnSOD level in proliferation as well as in invasive and angiogenic abilities of breast tumor cells by controlling superoxide anion radical (O2(•-)) and hydrogen peroxide (H2O2). Particularly, they report how these reactive oxygen species may activate some signaling pathways involved in breast tumor growth. Emerging understanding of these findings provides an interesting framework for guiding translational research and suggests a way to define precisely the clinical interest of MnSOD as a prognostic and/or predicting marker in breast cancer, by associating with some regulators involved in SOD2 gene regulation and other well-known biomarkers, in addition to the typical clinical parameters.
Collapse
Affiliation(s)
- Philippe Becuwe
- Centre de Recherche en Automatique de Nancy, UMR 7039 CNRS, Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy Cedex, France.
| | - Marie Ennen
- Centre de Recherche en Automatique de Nancy, UMR 7039 CNRS, Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy Cedex, France
| | - Rémi Klotz
- Centre de Recherche en Automatique de Nancy, UMR 7039 CNRS, Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy Cedex, France
| | - Claire Barbieux
- Centre de Recherche en Automatique de Nancy, UMR 7039 CNRS, Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy Cedex, France
| | - Stéphanie Grandemange
- Centre de Recherche en Automatique de Nancy, UMR 7039 CNRS, Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy Cedex, France
| |
Collapse
|
136
|
Ko YG, Hwang S, Kim SW, Kim H, Seong HH, Kim JH, Song Y, Yang BS, Song YM, Cho JH. Proteomic analysis of the extraembryonic tissues from cloned porcine fetus at day 35 of pregnancy. BMC Res Notes 2014; 7:861. [PMID: 25433481 PMCID: PMC4289280 DOI: 10.1186/1756-0500-7-861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 11/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Somatic cell cloning by nuclear transfer (SCNT) in pig is clearly of great benefit for basic research and biomedical applications. Even though cloned offspring have been successfully produced in pig, SCNT is struggling with the low efficiency. RESULTS In the present study, we investigated differentially expressed proteins of the extraembryonic tissue from pig SCNT fetus compared to control (normal) fetus. We obtained the extraembryonic tissue from embryos at day 35 of pregnancy and examined the protein expression profiles using two-dimensional electrophoresis (2-D) and Western blotting. The extraembryonic tissue of fetus in control pregnancy was compared to the extraembryonic tissue of SCNT fetus, which showed an abnormally small size and shape as well as exhibited abnormal placental morphology compared to control fetus. A proteomic analysis showed that the expression of 33 proteins was significantly increased or decreased in the extraembryonic tissue of SCNT fetus compared to control fetus. The differentially expressed proteins in the extraembryonic tissue of SCNT fetus included ATP or lipid binding proteins, antioxidant proteins, translation elongation factors, and transcription factors. Western blotting analysis indicated that antioxidant enzymes and anti-apoptotic proteins were down-regulated; however, the expression levels of apoptotic proteins, Bax and Hsp27, were increased in the extraembryonic tissue of SCNT fetus. Moreover, immunohistochemical analysis also showed that the expression of the catalase or GPX genes was decreased in the extraembryonic tissue with SCNT fetus compared to those with control fetus. In addition, we observed a significant decrease in DNA methytransferase1 (Dnmt1) expression in SCNT extraembryonic tissue, and the expression levels of Dnmt3a and Dnmt3b were abnormally higher in SCNT fetus compared to control fetus. Moreover, a marked increase in the frequency of TUNEL-positive cells was observed in the extraembryonic tissue in SCNT fetus. CONCLUSION These results demonstrated that pig SCNT fetus showed abnormal protein expression in the extraembryonic tissue, and extensive apoptosis occurred in the extraembryonic tissue of the SCNT fetus due to an increase in apoptotic protein expression or a decrease in antioxidant protein expression.
Collapse
Affiliation(s)
- Yeoung-Gyu Ko
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Effect of Uncaria tomentosa Extract on Apoptosis Triggered by Oxaliplatin Exposure on HT29 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:274786. [PMID: 25505920 PMCID: PMC4244697 DOI: 10.1155/2014/274786] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/17/2014] [Indexed: 02/08/2023]
Abstract
Background/Aim. The use of herbal products as a supplement to minimize the effects of chemotherapy for cancer treatment requires further attention with respect to the activity and toxicity of chemotherapy. Uncaria tomentosa extract, which contains oxindole alkaloids, is one of these herbal products. The objective of this study was to evaluate whether Uncaria tomentosa extract modulates apoptosis induced by chemotherapy exposure. Materials and Methods. Colorectal adenocarcinoma cells (HT29 cells) were grown in the presence of oxaliplatin and/or Uncaria tomentosa extract. Results. The hydroalcoholic extract of Uncaria tomentosa enhanced chemotherapy-induced apoptosis, with an increase in the percentage of Annexin positive cells, an increase in caspase activities, and an increase of DNA fragments in culture of the neoplastic cells. Moreover, antioxidant activity may be related to apoptosis. Conclusion. Uncaria tomentosa extract has a role for cancer patients as a complementary therapy. Further studies evaluating these beneficial effects with other chemotherapy drugs are recommended.
Collapse
|
138
|
Wu RM, Sun YY, Zhou TT, Zhu ZY, Zhuang JJ, Tang X, Chen J, Hu LH, Shen X. Arctigenin enhances swimming endurance of sedentary rats partially by regulation of antioxidant pathways. Acta Pharmacol Sin 2014; 35:1274-84. [PMID: 25152028 PMCID: PMC4186987 DOI: 10.1038/aps.2014.70] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/09/2014] [Indexed: 01/01/2023]
Abstract
AIM Arctigenin, a phenylpropanoid dibenzylbutyrolactone lignan found in traditional Chinese herbs, has been determined to exhibit a variety of pharmacological activities, including anti-tumor, anti-inflammation, neuroprotection, and endurance enhancement. In the present study, we investigated the antioxidation and anti-fatigue effects of arctigenin in rats. METHODS Rat L6 skeletal muscle cell line was exposed to H2O2 (700 μmol/L), and ROS level was assayed using DCFH-DA as a probe. Male SD rats were injected with arctigenin (15 mg·kg(-1)·d(-1), ip) for 6 weeks, and then the weight-loaded forced swimming test (WFST) was performed to evaluate their endurance. The levels of antioxidant-related genes in L6 cells and the skeletal muscles of rats were analyzed using real-time RT-PCR and Western blotting. RESULTS Incubation of L6 cells with arctigenin (1, 5, 20 μmol/L) dose-dependently decreased the H2O2-induced ROS production. WFST results demonstrated that chronic administration of arctigenin significantly enhanced the endurance of rats. Furthermore, molecular biology studies on L6 cells and skeletal muscles of the rats showed that arctigenin effectively increased the expression of the antioxidant-related genes, including superoxide dismutase (SOD), glutathione reductase (Gsr), glutathione peroxidase (GPX1), thioredoxin (Txn) and uncoupling protein 2 (UCP2), through regulation of two potential antioxidant pathways: AMPK/PGC-1α/PPARα in mitochondria and AMPK/p53/Nrf2 in the cell nucleus. CONCLUSION Arctigenin efficiently enhances rat swimming endurance by elevation of the antioxidant capacity of the skeletal muscles, which has thereby highlighted the potential of this natural product as an antioxidant in the treatment of fatigue and related diseases.
Collapse
Affiliation(s)
- Ruo-ming Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-yan Sun
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ting-ting Zhou
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhi-yuan Zhu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing-jing Zhuang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xuan Tang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Chen
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Li-hong Hu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xu Shen
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
139
|
Kim DH, Song NY, Kim EH, Na HK, Joe Y, Chung HT, Surh YJ. 15-Deoxy-Δ12,14-prostaglandin J2induces p53 expression through Nrf2-mediated upregulation of heme oxygenase-1 in human breast cancer cells. Free Radic Res 2014; 48:1018-27. [DOI: 10.3109/10715762.2014.897343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
140
|
Thakur S, Sarkar B, Cholia RP, Gautam N, Dhiman M, Mantha AK. APE1/Ref-1 as an emerging therapeutic target for various human diseases: phytochemical modulation of its functions. Exp Mol Med 2014; 46:e106. [PMID: 25033834 PMCID: PMC4119211 DOI: 10.1038/emm.2014.42] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/27/2014] [Accepted: 03/05/2014] [Indexed: 12/12/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional enzyme involved in the base excision repair (BER) pathway, which repairs oxidative base damage caused by endogenous and exogenous agents. APE1 acts as a reductive activator of many transcription factors (TFs) and has also been named redox effector factor 1, Ref-1. For example, APE1 activates activator protein-1, nuclear factor kappa B, hypoxia-inducible factor 1α, paired box gene 8, signal transducer activator of transcription 3 and p53, which are involved in apoptosis, inflammation, angiogenesis and survival pathways. APE1/Ref-1 maintains cellular homeostasis (redox) via the activation of TFs that regulate various physiological processes and that crosstalk with redox balancing agents (for example, thioredoxin, catalase and superoxide dismutase) by controlling levels of reactive oxygen and nitrogen species. The efficiency of APE1/Ref-1's function(s) depends on pairwise interaction with participant protein(s), the functions regulated by APE1/Ref-1 include the BER pathway, TFs, energy metabolism, cytoskeletal elements and stress-dependent responses. Thus, APE1/Ref-1 acts as a ‘hub-protein' that controls pathways that are important for cell survival. In this review, we will discuss APE1/Ref-1's versatile nature in various human etiologies, including neurodegeneration, cancer, cardiovascular and other diseases that have been linked with alterations in the expression, subcellular localization and activities of APE/Ref-1. APE1/Ref-1 can be targeted for therapeutic intervention using natural plant products that modulate the expression and functions of APE1/Ref-1. In addition, studies focusing on translational applications based on APE1/Ref-1-mediated therapeutic interventions are discussed.
Collapse
Affiliation(s)
- Shweta Thakur
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Bibekananda Sarkar
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Ravi P Cholia
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Nandini Gautam
- Center for Environmental Science and Technology, School of Environment and Earth Sciences, Central University of Punjab, Punjab, India
| | - Monisha Dhiman
- Center for Genetic Diseases and Molecular Medicine, School of Emerging Life Science Technologies, Central University of Punjab, Punjab, India
| | - Anil K Mantha
- 1] Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India [2] Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
141
|
Abstract
SIGNIFICANCE Oxidative (reactive oxygen species [ROS]) and nitrosative (reactive nitrogen species [RNS]) stress affects many physiological processes, including survival and death. Although high levels of ROS/RNS mainly causes cell death, low levels of free radicals directly modulate the activities of transcriptional factors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), p53, and nuclear factor (erythroid-derived) 2-like (Nrf2), and regulate numerous protein kinase cascades that participate in the regulation of the cross talk between autophagy and apoptosis. RECENT ADVANCES Low levels of ROS modify Atg4 and high mobility group box 1 (HMGB1) proteins, activate AMP-activated protein kinase (AMPK) and apoptosis signal-regulating kinase/c-Jun N-terminal kinase (JNK) pathways, or transactivate various proteins that could upregulate autophagy, leading to reductions in apoptosis. Transactivation of antioxidant genes blocks apoptosis and serves as a feedback loop to reduce autophagy. Free radicals could also activate protein kinase B (PKB, or Akt), preventing both autophagy and apoptosis. Stimulation of nitric oxide formation causes S-nitrosylation of several kinases, including JNK1 and IκB kinase β, which blocks autophagy and could promote apoptosis. However, S-nitrosylation of some proapoptotic proteins could block apoptosis. CRITICAL ISSUES Endoplasmic reticulum and mitochondria are the main sources of free radicals, which play an essential role in the regulation of apoptosis and autophagy. Oxidation of cardiolipin promotes cytochrome c release and apoptosis that potentially could be inhibited by autophagic clearance of damaged mitochondria. Elimination of damaged mitochondria reduces ROS accumulation, creating a feedback loop that causes inhibition of autophagy. Low levels of RNS could inhibit fission of mitochondria, which would block their degradation by autophagy and spare cells from apoptosis. FUTURE DIRECTIONS Understanding of mechanisms that regulate the cross talk between cell fates is essential for discovery of therapeutic tools in the strenuous fight against various disorders, including neurodegeneration and cancer.
Collapse
Affiliation(s)
- Vitaliy O Kaminskyy
- 1 Division of Toxicology, Institute of Environmental Medicine , Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
142
|
Ureshino RP, Rocha KK, Lopes GS, Bincoletto C, Smaili SS. Calcium signaling alterations, oxidative stress, and autophagy in aging. Antioxid Redox Signal 2014; 21:123-37. [PMID: 24512092 DOI: 10.1089/ars.2013.5777] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Aging is a multi-factorial process that may be associated with several functional and structural deficits which can evolve into degenerative diseases. In this review, we present data that may depict an expanded view of molecular aging theories, beginning with the idea that reactive oxygen species (ROS) are the major effectors in this process. In addition, we have correlated the importance of autophagy as a neuroprotective mechanism and discussed a link between age-related molecules, Ca(2+) signaling, and oxidative stress. RECENT ADVANCES There is evidence suggesting that alterations in Ca(2+) homeostasis, including mitochondrial Ca(2+) overload and alterations in electron transport chain (ETC) complexes, which increase cell vulnerability, are linked to oxidative stress in aging. As much as Ca(2+) signaling is altered in aged cells, excess ROS can be produced due to an ineffective coupling of mitochondrial respiration. Damaged mitochondria might not be removed by the macroautophagic system, which is hampered in aging by lipofuscin accumulation, boosting ROS generation, damaging DNA, and, ultimately, leading to apoptosis. CRITICAL ISSUES This process can lead to altered protein expression (such as p53, Sirt1, and IGF-1) and progress to cell death. This cycle can lead to increased cell vulnerability in aging and contribute to an increased susceptibility to degenerative processes. FUTURE DIRECTIONS A better understanding of Ca(2+) signaling and molecular aging alterations is important for preventing apoptosis in age-related diseases. In addition, caloric restriction, resveratrol and autophagy modulation appear to be predominantly cytoprotective, and further studies of this process are promising in age-related disease therapeutics.
Collapse
|
143
|
Zhu X, Zheng X, Wu Y. Cleaved high molecular weight kininogen stimulates JNK/FOXO4/MnSOD pathway for induction of endothelial progenitor cell senescence. Biochem Biophys Res Commun 2014; 450:1261-5. [PMID: 24984152 DOI: 10.1016/j.bbrc.2014.06.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/23/2014] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Recently we have reported that cleaved high molecular weight kininogen (HKa) accelerates the onset of endothelial progenitor cells (EPCs) senescence by induction of reactive oxygen species (ROS). However, the mechanisms by which HKa induces production of ROS remain unknown. In this study, we have shown that HKa induces EPC senescence via stimulation of c-Jun N-terminal kinases (JNK)-related pathway. METHODS AND RESULTS Treatment of human EPCs with HKa for 72h stimulated JNK phosphorylation at Thr183/Tyr185, and FOXO4 phosphorylation at Thr451, Concomitantly, upregulated the expression of MnSOD at protein and mRNA levels in a concentration-dependent manner. HKa increased intracellular level of H2O2, without affecting the expression of catalase. To narrow down the functional domain of HKa, recombinant proteins of human HK heavy chain (HC, 19-380aa) and light chain (LC, 390-644aa) were generated. HC, but not LC, increased senescence of EPCs and intracellular ROS levels, to a similar extent with HKa. Moreover, HC at 50 nM increased FOXO4 phosphorylation at Thr451 and the protein level of MnSOD in EPCs. CONCLUSION These results demonstrate that HKa accelerates the onset of EPC senescence by stimulating JNK/FOXO4/MnSOD pathway, its effect is mediated by the HC.
Collapse
Affiliation(s)
- Xuemei Zhu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xichen Zheng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yi Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
144
|
El-Zahany EA, Ali MM, Drweesh SA, El-Seidy AMA, Abdel-Wahab BF, Youssef NS. Synthesis, Characterization, and Antiproliferative Activity of Cu2+, V(IV)O2+, Co2+, Mn2+, and Ni2+ Complexes with 3-(2-(4-Methoxyphenylcarbamothioyl)Hydrazinyl)-3-OXO-N-(Thiazol-2-yl)Propanamide against Human Breast Adenocarcinoma Cells. PHOSPHORUS SULFUR 2014. [DOI: 10.1080/10426507.2013.855764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Eman A. El-Zahany
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Mamdouh M. Ali
- Biochemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Sayed A. Drweesh
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Ahmed M. A. El-Seidy
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Bakr F. Abdel-Wahab
- Applied Organic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Nabil S. Youssef
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| |
Collapse
|
145
|
Ismail AFM, Ali MM, Ismail LFM. Photodynamic therapy mediated antiproliferative activity of some metal-doped ZnO nanoparticles in human liver adenocarcinoma HepG2 cells under UV irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 138:99-108. [PMID: 24911277 DOI: 10.1016/j.jphotobiol.2014.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/29/2014] [Accepted: 04/09/2014] [Indexed: 01/10/2023]
Abstract
Photodynamic therapy (PDT) is a promising new modality for the treatment of cancer through generation of reactive oxygen species (ROS). In this work, human liver adenocarcinoma cells HepG2 were treated with zinc oxide nanoparticles (ZnO-NPs), metal-doped-ZnO-NPs: Fe-ZnO-NPs Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs, Silica-coated ZnO-NPs, titanium dioxide nanoparticles (TiO2-NPs), titanium dioxide nano-tubes (TiO2-NTs) and ZnO-NPs/TiO2-NTs nanocomposite under UV irradiation. Doxorubicin was used as a standard drug. The results demonstrated that the ZnO-NPs, Fe-ZnO-NPs, Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs showed cytotoxicity against HepG2 cells, with the median growth inhibitory concentrations (IC50) 42.60, 37.20, 45.10, 77.20 and 56.50 μg/ml, respectively, as compared to doxorubicin (IC50: 20.10 μg/ml). Treatment of the cancer cells with ZnO-NPs, Fe-ZnO-NPs, Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs resulted in a significant increase in the activity of SOD and the levels of H2O2 and NO than those of control, accompanied with a significant decrease in the activity of CAT and GSH-Px. Also, depletion of reduced GSH, total protein and nucleic acids levels was observed. In conclusion, metal-doped ZnO-NPs may induce antiproliferative effect on HepG2 cells under UV-irradiation due to generation of ROS. Therefore, they could be included in modern clinical trials after in vivo more investigations, using photodynamic therapy technique.
Collapse
Affiliation(s)
- Amel F M Ismail
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Nasr City, Cairo, Egypt.
| | - Mamdouh M Ali
- Biochemistry Department, Division of Genetic Engineering and Biotechnology, National Research Centre, Dokki, Giza, Egypt
| | - Laila F M Ismail
- Chemistry Department, Al-Azhar University, Faculty of Science, Nasr City, Cairo, Egypt
| |
Collapse
|
146
|
Non-thermal atmospheric pressure plasma preferentially induces apoptosis in p53-mutated cancer cells by activating ROS stress-response pathways. PLoS One 2014; 9:e91947. [PMID: 24759730 PMCID: PMC3997341 DOI: 10.1371/journal.pone.0091947] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/18/2014] [Indexed: 01/26/2023] Open
Abstract
Non-thermal atmospheric pressure plasma (NTAPP) is an ionized gas at room temperature and has potential as a new apoptosis-promoting cancer therapy that acts by generating reactive oxygen species (ROS). However, it is imperative to determine its selectivity and standardize the components and composition of NTAPP. Here, we designed an NTAPP-generating apparatus combined with a He gas feeding system and demonstrated its high selectivity toward p53-mutated cancer cells. We first determined the proper conditions for NTAPP exposure to selectively induce apoptosis in cancer cells. The apoptotic effect of NTAPP was greater for p53-mutated cancer cells; artificial p53 expression in p53-negative HT29 cells decreased the pro-apoptotic effect of NTAPP. We also examined extra- and intracellular ROS levels in NTAPP-treated cells to deduce the mechanism of NTAPP action. While NTAPP-mediated increases in extracellular nitric oxide (NO) did not affect cell viability, intracellular ROS increased under NTAPP exposure and induced apoptotic cell death. This effect was dose-dependently reduced following treatment with ROS scavengers. NTAPP induced apoptosis even in doxorubicin-resistant cancer cell lines, demonstrating the feasibility of NTAPP as a potent cancer therapy. Collectively, these results strongly support the potential of NTAPP as a selective anticancer treatment, especially for p53-mutated cancer cells.
Collapse
|
147
|
Candas D, Li JJ. MnSOD in oxidative stress response-potential regulation via mitochondrial protein influx. Antioxid Redox Signal 2014; 20:1599-617. [PMID: 23581847 PMCID: PMC3942709 DOI: 10.1089/ars.2013.5305] [Citation(s) in RCA: 473] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE The mitochondrial antioxidant manganese superoxide dismutase (MnSOD) is encoded by genomic DNA and its dismutase function is fully activated in the mitochondria to detoxify free radical O2(•-) generated by mitochondrial respiration. Accumulating evidence shows an extensive communication between the mitochondria and cytoplasm under oxidative stress. Not only is the MnSOD gene upregulated by oxidative stress, but MnSOD activity can be enhanced via the mitochondrial protein influx (MPI). RECENT ADVANCES A cluster of MPI containing cytoplasmic/nuclear proteins, such as cyclins, cyclin-dependent kinases, and p53 interact with and alter MnSOD activity. These proteins modulate MnSOD superoxide scavenging activity via post-translational modifications in the mitochondria. In addition to well-established pathways in gene expression, recent findings suggest that MnSOD enzymatic activity can also be enhanced by phosphorylation of specific motifs in mitochondria. This review attempts to discuss the pre- and post-translational regulation of MnSOD, and how these modifications alter MnSOD activity, which induces a cell adaptive response to oxidative stress. CRITICAL ISSUES MnSOD is biologically significant to aerobic cells. Its role in protecting the cells against the deleterious effects of reactive oxygen species is evident. However, the exact network of MnSOD-associated cellular adaptive reaction to oxidative stress and its post-translational modifications, especially its enzymatic enhancement via phosphorylation, is not yet fully understood. FUTURE DIRECTIONS The broad discussion of the multiple aspects of MnSOD regulation, including gene expression, protein modifications, and enzymatic activity, will shed light onto the unknown mechanisms that govern the prosurvival networks involved in cellular and mitochondrial adaptive response to genotoxic environment.
Collapse
Affiliation(s)
- Demet Candas
- 1 Department of Radiation Oncology, University of California Davis , Sacramento, California
| | | |
Collapse
|
148
|
Wang F, Franco R, Skotak M, Hu G, Chandra N. Mechanical stretch exacerbates the cell death in SH-SY5Y cells exposed to paraquat: mitochondrial dysfunction and oxidative stress. Neurotoxicology 2014; 41:54-63. [PMID: 24462953 DOI: 10.1016/j.neuro.2014.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/03/2013] [Accepted: 01/12/2014] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that traumatic brain injury (TBI) and pesticide exposure increase the risk of Parkinson's disease (PD), but the molecular mechanisms involved remain unclear. Using an in vitro model of TBI, we evaluated the role of mitochondrial membrane potential (ΔΨm) and mitochondrial reactive oxygen species (ROS) induced by stretch on dopaminergic cell death upon paraquat exposure. Human dopaminergic neuroblastoma SH-SY5Y cells grown on silicone membrane were stretched at mild (25%) and moderate (50%) strain prior to paraquat exposure. We observed that moderate stretch (50% strain) increased the vulnerability of cells to paraquat demonstrated by the loss of plasma membrane integrity (propidium iodide-uptake) and decreased mitochondrial activity (MTT assay). Mitochondrial depolarization occurred immediately after stretch, while mitochondrial ROS increased rapidly and remained elevated for up to 4h after the stretch injury. Intracellular glutathione (GSH) stores were also transiently decreased immediately after moderate stretch. Cells treated with paraquat, or moderate stretch exhibited negligible mitochondrial depolarization at 48h post treatment, whereas in cells stretched prior to paraquat exposure, a significant mitochondrial depolarization occurred compared to samples exposed to either paraquat or stretch. Moderate stretch also increased mitochondrial ROS formation, as well as exacerbated intracellular GSH loss induced by paraquat. Overexpression of manganese superoxide dismutase (MnSOD) markedly diminished the deleterious effects of stretch in paraquat neurotoxicity. Our findings demonstrate that oxidative stress induced by mitochondrial dysfunction plays a critical role in the synergistic toxic effects of stretch (TBI) and pesticide exposure. Mitigation of oxidative stress via mitochondria-targeted antioxidants appears an attractive route for treatment of neurodegeneration mediated by TBI.
Collapse
Affiliation(s)
- Fang Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588-0526, USA
| | - Rodrigo Franco
- Redox Biology Center & School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588-0526, USA
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Namas Chandra
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102-1982, USA.
| |
Collapse
|
149
|
Wang DB, Kinoshita C, Kinoshita Y, Morrison RS. p53 and mitochondrial function in neurons. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1186-97. [PMID: 24412988 DOI: 10.1016/j.bbadis.2013.12.015] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/24/2013] [Accepted: 12/28/2013] [Indexed: 01/08/2023]
Abstract
The p53 tumor suppressor plays a central role in dictating cell survival and death as a cellular sensor for a myriad of stresses including DNA damage, oxidative and nutritional stress, ischemia and disruption of nucleolar function. Activation of p53-dependent apoptosis leads to mitochondrial apoptotic changes via the intrinsic and extrinsic pathways triggering cell death execution most notably by release of cytochrome c and activation of the caspase cascade. Although it was previously believed that p53 induces apoptotic mitochondrial changes exclusively through transcription-dependent mechanisms, recent studies suggest that p53 also regulates apoptosis via a transcription-independent action at the mitochondria. Recent evidence further suggests that p53 can regulate necrotic cell death and autophagic activity including mitophagy. An increasing number of cytosolic and mitochondrial proteins involved in mitochondrial metabolism and respiration are regulated by p53, which influences mitochondrial ROS production as well. Cellular redox homeostasis is also directly regulated by p53 through modified expression of pro- and anti-oxidant proteins. Proper regulation of mitochondrial size and shape through fission and fusion assures optimal mitochondrial bioenergetic function while enabling adequate mitochondrial transport to accommodate local energy demands unique to neuronal architecture. Abnormal regulation of mitochondrial dynamics has been increasingly implicated in neurodegeneration, where elevated levels of p53 may have a direct contribution as the expression of some fission/fusion proteins are directly regulated by p53. Thus, p53 may have a much wider influence on mitochondrial integrity and function than one would expect from its well-established ability to transcriptionally induce mitochondrial apoptosis. However, much of the evidence demonstrating that p53 can influence mitochondria through nuclear, cytosolic or intra-mitochondrial sites of action has yet to be confirmed in neurons. Nonetheless, as mitochondria are essential for supporting normal neuronal functions and in initiating/propagating cell death signaling, it appears certain that the mitochondria-related functions of p53 will have broader implications than previously thought in acute and progressive neurological conditions, providing new therapeutic targets for treatment.
Collapse
Affiliation(s)
- David B Wang
- Department of Neurological Surgery, University of Washington School of Medicine, Box 356470, Seattle, WA 98195-6470, USA
| | - Chizuru Kinoshita
- Department of Neurological Surgery, University of Washington School of Medicine, Box 356470, Seattle, WA 98195-6470, USA
| | - Yoshito Kinoshita
- Department of Neurological Surgery, University of Washington School of Medicine, Box 356470, Seattle, WA 98195-6470, USA
| | - Richard S Morrison
- Department of Neurological Surgery, University of Washington School of Medicine, Box 356470, Seattle, WA 98195-6470, USA.
| |
Collapse
|
150
|
Abstract
Cancers cells shift their metabolism towards glycolysis in order to help them support the biosynthetic demands necessary to sustain cell proliferation and growth, adapt to stress and avoid excessive reactive oxygen species (ROS) accumulation. While the p53 tumor suppressor protein is known to inhibit cell growth by inducing apoptosis, senescence and cell cycle arrest, recent studies have found that p53 is also able to influence cell metabolism. TIGAR is a p53 target that functions as a fructose-2,6-bisphosphatase, thereby lowering glycolytic flux and promoting antioxidant functions. By protecting cells from oxidative stress, TIGAR may mediate some of the tumor suppressor activity of p53 but could also contribute to tumorigenesis. Here we discuss the activities of TIGAR described so far, and the potential consequences of TIGAR expression on normal and tumor cells.
Collapse
Affiliation(s)
- Pearl Lee
- Cancer Research-UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK
| | - Karen H Vousden
- Cancer Research-UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK
| | - Eric C Cheung
- Cancer Research-UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK
| |
Collapse
|