1
|
Wydorski PJ, Zmijewska A, Franczak A. The Extremely-Low-Frequency Electromagnetic Field Affects Apoptosis and Oxidative-Stress-Related Genes and Proteins in the Porcine Endometrium-An In Vitro Study. Int J Mol Sci 2024; 25:6931. [PMID: 39000040 PMCID: PMC11241303 DOI: 10.3390/ijms25136931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Nowadays, the extremely-low-frequency electromagnetic field (ELF-EMF) is recognized as environmental pollution. The data indicate that the ELF-EMF may affect factors related to epigenetic regulation and alter important biological processes in the uterus. The impact of the ELF-EMF on apoptosis and oxidative-stress-related genes has not been documented in porcine endometrium. This raises the question of whether the exposure to the ELF-EMF can induce apoptosis and/or oxidative stress in the endometrium of pigs during the peri-implantation period. Porcine endometrial slices (100 ± 5 mg) collected (n = 5) during the peri-implantation period were treated in vitro with ELF-EMF at a frequency of 50 Hz and flux density of 8 × 104 mG for 2 h. To determine the effect of ELF-EMF on apoptosis and oxidative stress in the endometrium, CASP3, CASP7, CIDEB, GADD45G, NOS1, NOS2, NOS3, and TP53I3 mRNA transcript were analyzed using real-time PCR, and protein abundance of CASP3, CASP7 using Western blot, and eNOS using ELISA were determined. Moreover, CASP3/7 and NOS activity was analyzed using flow cytometry and colorimetry, respectively. The decreased CASP7 and increased NOS3 mRNA transcript and protein abundance in ELF-EMF-treated endometrium were observed. Moreover, CIDEB, GADD45G, and TP53I3 mRNA transcript abundance was increased. Only p ≤ 0.05 was considered a statistically significant difference. The documented alterations indicate the potential of the ELF-EMF to affect apoptosis and generate oxidative stress in the endometrium. The insight into observed consequences documents for the first time the fact that the ELF-EMF may influence endometrial cell proliferation, angiogenesis, and/or tissue receptivity during peri-implantation.
Collapse
Affiliation(s)
| | | | - Anita Franczak
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland; (P.J.W.); (A.Z.)
| |
Collapse
|
2
|
Yan L, Li S, Hu Q, Liao D. Genetic correlations, shared risk genes and immunity landscapes between COVID-19 and venous thromboembolism: evidence from GWAS and bulk transcriptome data. Inflamm Res 2024:10.1007/s00011-024-01857-w. [PMID: 38433131 DOI: 10.1007/s00011-024-01857-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/20/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Patients with coronavirus disease 2019 (COVID-19) were vulnerable to venous thromboembolism (VTE), which further increases the risk of unfavorable outcomes. However, neither genetic correlations nor shared genes underlying COVID-19 and VTE are well understood. OBJECTIVE This study aimed to characterize genetic correlations and common pathogenic mechanisms between COVID-19 and VTE. METHODS We used linkage disequilibrium score (LDSC) regression and Mendelian Randomization (MR) analysis to investigate the genetic associations and causal effects between COVID-19 and VTE, respectively. Then, the COVID-19 and VTE-related datasets were obtained from the Gene Expression Omnibus (GEO) database and analyzed by bioinformatics and systems biology approaches with R software, including weighted gene co-expression network analysis (WGCNA), enrichment analysis, and single-cell transcriptome sequencing analysis. The miRNA-genes and transcription factor (TF)-genes interaction networks were conducted by NetworkAnalyst. We performed the secondary analysis of the ATAC-seq and Chip-seq datasets to address the epigenetic-regulating relationship of the shared genes. RESULTS This study demonstrated positive correlations between VTE and COVID-19 by LDSC and bidirectional MR analysis. A total of 26 potential shared genes were discovered from the COVID-19 dataset (GSE196822) and the VTE dataset (GSE19151), with 19 genes showing positive associations and 7 genes exhibiting negative associations with these diseases. After incorporating two additional datasets, GSE164805 (COVID-19) and GSE48000 (VTE), two hub genes TP53I3 and SLPI were identified and showed up-regulation and diagnostic capabilities in both illnesses. Furthermore, this study illustrated the landscapes of immune processes in COVID-19 and VTE, revealing the downregulation in effector memory CD8+ T cells and activated B cells. The single-cell sequencing analysis suggested that the hub genes were predominantly expressed in the monocytes of COVID-19 patients at high levels. Additionally, we identified common regulators of hub genes, including five miRNAs (miR-1-3p, miR-203a-3p, miR-210-3p, miR-603, and miR-124-3p) and one transcription factor (RELA). CONCLUSIONS Collectively, our results highlighted the significant correlations between COVID-19 and VTE and pinpointed TP53I3 and SLPI as hub genes that potentially link the severity of both conditions. The hub genes and their common regulators might present an opportunity for the simultaneous treatment of these two diseases.
Collapse
Affiliation(s)
- Langchao Yan
- Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, Shanxi, China
| | - Shifu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Street, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, 87 Xiangya Street, Changsha, 410008, Hunan, China
| | - Qian Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Di Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, 87 Xiangya Street, Changsha, 410008, Hunan, China.
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Street, Changsha, 410008, Hunan, China.
| |
Collapse
|
3
|
Ray SK, Jayashankar E, Kotnis A, Mukherjee S. Oxidative versus Reductive Stress in Breast Cancer Development and Cellular Mechanism of Alleviation: A Current Perspective with Anti-breast Cancer Drug Resistance. Curr Mol Med 2024; 24:205-216. [PMID: 36892117 DOI: 10.2174/1566524023666230309112751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 03/10/2023]
Abstract
Redox homeostasis is essential for keeping our bodies healthy, but it also helps breast cancer cells grow, stay alive, and resist treatment. Changes in the redox balance and problems with redox signaling can make breast cancer cells grow and spread and make them resistant to chemotherapy and radiation therapy. Reactive oxygen species/reactive nitrogen species (ROS/RNS) generation and the oxidant defense system are out of equilibrium, which causes oxidative stress. Many studies have shown that oxidative stress can affect the start and spread of cancer by interfering with redox (reduction-oxidation) signaling and damaging molecules. The oxidation of invariant cysteine residues in FNIP1 is reversed by reductive stress, which is brought on by protracted antioxidant signaling or mitochondrial inactivity. This permits CUL2FEM1B to recognize its intended target. After the proteasome breaks down FNIP1, mitochondrial function is restored to keep redox balance and cell integrity. Reductive stress is caused by unchecked amplification of antioxidant signaling, and changes in metabolic pathways are a big part of breast tumors' growth. Also, redox reactions make pathways like PI3K, PKC, and protein kinases of the MAPK cascade work better. Kinases and phosphatases control the phosphorylation status of transcription factors like APE1/Ref-1, HIF-1, AP-1, Nrf2, NF-B, p53, FOXO, STAT, and - catenin. Also, how well anti-breast cancer drugs, especially those that cause cytotoxicity by making ROS, treat patients depends on how well the elements that support a cell's redox environment work together. Even though chemotherapy aims to kill cancer cells, which it does by making ROS, this can lead to drug resistance in the long run. The development of novel therapeutic approaches for treating breast cancer will be facilitated by a better understanding of the reductive stress and metabolic pathways in tumor microenvironments.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh, 462020, India
| | - Erukkambattu Jayashankar
- Department of Pathology & Lab Medicine, All India Institute of Medical Sciences-Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India
| | - Ashwin Kotnis
- Department of Biochemistry, All India Institute of Medical Sciences-Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences-Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India
| |
Collapse
|
4
|
Zhang G, Zhang Y, Jing L, Zhao H. Lead exposure induced developmental nephrotoxicity in Japanese quail (Coturnix japonica) via oxidative stress-based PI3K/AKT pathway inhibition and NF-κB pathway activation. Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109599. [PMID: 36893933 DOI: 10.1016/j.cbpc.2023.109599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
Birds are sensitive to environmental pollution and lead (Pb) contamination could negatively affect nearly all avian organs and systems including kidney of excretive system. Thereby, we used a biological model species-Japanese quail (Coturnix japonica) to examine the nephrotoxic effects of Pb exposure and possible toxic mechanism of Pb on birds. Quail chicks of 7-day-old were exposed to 50 ppm Pb of low dose and high dose of 500 ppm and 1000 ppm Pb in drinking water for five weeks. The results showed that Pb exposure induced kidney weight increase while body weight and length reduction. The increase of uric acid (UA), creatinine (CREA) and cystatin c (Cys C) in the plasma suggested renal dysfunction. Moreover, both microstructural and ultrastructural changes demonstrated obvious kidney damages. In particular, renal tubule epithelial cells and glomeruli swelling indicated renal inflammation. Furthermore, changes in the content and activity of oxidative stress markers suggested that Pb caused excessive oxidative stress in the kidney. Pb exposure also induced abnormal apoptosis in the kidney. In addition, RNA sequencing (RNA-Seq) analysis revealed that Pb disturbed molecular pathways and signaling related with renal function. Especially, Pb exposure resulted in an increase in renal uric acid synthesis by disrupting purine metabolism. Pb caused apoptotic increment by inhibiting the phosphatidylinositol-3-kinase (PI3K)/RAC-alpha serine/threonine-protein kinase (AKT) pathway and induced aggravated inflammation by activating Nuclear Factor kappa B (NF-κB) signaling pathway. The study implied that Pb caused nephrotoxicity through structural damages, uric acid metabolism disorder, oxidation imbalance, apoptosis and inflammatory pathway activation.
Collapse
Affiliation(s)
- Gaixia Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yuxin Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Lingyang Jing
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
5
|
Zhou H, Tang L, Shang ZF, Zhou PK, Li M. PIG3 downregulation enhances the radio sensitivity of NSCLC cells by promoting G2/M cell cycle arrest and apoptosis. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
6
|
Guo W, Liang D, Wang P, Yin L, Zhang H, Xing C, Huang Z, Wu Y, Li H, Cheng Z, Xiao X, Liu J, Wang Z, Peng H. HIF-PH Encoded by EGLN1 Is a Potential Therapeutic Target for Chronic Lymphocytic Leukemia. Pharmaceuticals (Basel) 2022; 15:ph15060734. [PMID: 35745653 PMCID: PMC9229586 DOI: 10.3390/ph15060734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Owing to the recent emergence of drug resistance to Bruton's tyrosine kinase inhibitors (BTK) in chronic lymphocytic leukemia (CLL) treatment, it is crucial to identify alternative therapeutic targets. Therefore, we aimed to identify therapeutic options for CLL besides BTK. We identified that HIF1A expression was higher in CLL patients than in controls, which may suggest good prognosis. We used a lentiviral knockdown of EGLN1 (encoding hypoxia-inducible factor prolyl hydroxylase [HIF-PH]) and found that the growth of MEC-1 cells slowed in the knockdown group. Treatment of CLL cell lines MEC-1 and HG3 with the HIF-PH inhibitor molidustat showed that molidustat could induce apoptosis in a concentration-dependent manner in CLL cells and had low cytotoxicity at this concentration. CXCR4, HIF1A, SLC2AI, and VEGF, the downstream molecules of the HIF pathway, were upregulated after molidustat treatment. Western blotting results indicated that molidustat increased HIF1A expression in CLL cell lines and cells from CLL patients, and sequencing/quantitative PCR analysis demonstrated that the ribosome biogenesis pathway was inhibited in MEC-1 cells after molidustat treatment. We further identified synergistic cytotoxicity of molidustat in combination with ibrutinib on the MEC-1 and HG3 cell lines at certain concentrations. Therefore, molidustat is a potential therapeutic option for CLL.
Collapse
Affiliation(s)
- Wancheng Guo
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Daomiao Liang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China;
| | - Peilong Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Cheng Xing
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Yinghua Wu
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
| | - Xiaojuan Xiao
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Correspondence: (Z.W.); (H.P.); Tel.: +86-155-7588-6164 (Z.W.); +86-731-8529-5296 (H.P.)
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (P.W.); (L.Y.); (H.Z.); (C.X.); (Z.H.); (H.L.); (Z.C.); (X.X.); (J.L.)
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha 410011, China
- Correspondence: (Z.W.); (H.P.); Tel.: +86-155-7588-6164 (Z.W.); +86-731-8529-5296 (H.P.)
| |
Collapse
|
7
|
Liu S, Zhang Y, Qiu L, Zhang S, Meng Y, Huang C, Chen Z, Zhang B, Han J. Uncovering N4-Acetylcytidine-Related mRNA Modification Pattern and Landscape of Stemness and Immunity in Hepatocellular Carcinoma. Front Cell Dev Biol 2022; 10:861000. [PMID: 35493106 PMCID: PMC9046676 DOI: 10.3389/fcell.2022.861000] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/04/2022] [Indexed: 12/25/2022] Open
Abstract
N4-acetylcytidine (ac4C) is an ancient and conserved RNA modification. Previously, ac4C mRNA modification has been reported promoting proliferation and metastasis of tumor cells. However, it remains unclear whether and how ac4C-related mRNA modification patterns influencing the prognosis of hepatocellular carcinoma (HCC) patients. Hereby, we constructed an ac4Cscore model and classified patients into two groups and investigated the potential intrinsic and extrinsic characteristics of tumor. The ac4Cscore model, including COL15A1, G6PD and TP53I3, represented ac4C-related mRNA modification patterns in HCC. According to ac4Cscore, patients were stratified to high and low groups with distinct prognosis. Patients subject to high group was related to advanced tumor stage, higher TP53 mutation rate, higher tumor stemness, more activated pathways in DNA-repair system, lower stromal score, higher immune score and higher infiltrating of T cells regulatory. While patients attributed to low group were correlated with abundance of T cells CD4 memory, less aggressive immune subtype and durable therapy benefit. We also found ac4Cscore as a novel marker to predict patients’ prognosis with anti-PD1 immunotherapy and/or mTOR inhibitor treatment. Our study for the first time showed the association between ac4C-related mRNA modification patterns and tumor intrinsic and extrinsic characteristics, thus influencing the prognosis of patients.
Collapse
Affiliation(s)
- Sicheng Liu
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yaguang Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Qiu
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Su Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Meng
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Canhua Huang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhixin Chen
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastrointestinal Surgery, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastrointestinal Surgery, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Han
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Junhong Han,
| |
Collapse
|
8
|
Morelli AP, Tortelli TC, Mancini MCS, Pavan ICB, Silva LGS, Severino MB, Granato DC, Pestana NF, Ponte LGS, Peruca GF, Pauletti BA, Dos Santos DFG, de Moura LP, Bezerra RMN, Leme AFP, Chammas R, Simabuco FM. STAT3 contributes to cisplatin resistance, modulating EMT markers, and the mTOR signaling in lung adenocarcinoma. Neoplasia 2021; 23:1048-1058. [PMID: 34543857 PMCID: PMC8453219 DOI: 10.1016/j.neo.2021.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 12/29/2022]
Abstract
Lung cancer is the second leading cause of cancer death worldwide and is strongly associated with cisplatin resistance. The transcription factor signal transducer and activator of transcription 3 (STAT3) is constitutively activated in cancer cells and coordinates critical cellular processes as survival, self-renewal, and inflammation. In several types of cancer, STAT3 controls the development, immunogenicity, and malignant behavior of tumor cells while it dictates the responsiveness to radio- and chemotherapy. It is known that STAT3 phosphorylation at Ser727 by mechanistic target of rapamycin (mTOR) is necessary for its maximal activation, but the crosstalk between STAT3 and mTOR signaling in cisplatin resistance remains elusive. In this study, using a proteomic approach, we revealed important targets and signaling pathways altered in cisplatin-resistant A549 lung adenocarcinoma cells. STAT3 had increased expression in a resistance context, which can be associated with a poor prognosis. STAT3 knockout (SKO) resulted in a decreased mesenchymal phenotype in A549 cells, observed by clonogenic potential and by the expression of epithelial-mesenchymal transition markers. Importantly, SKO cells did not acquire the mTOR pathway overactivation induced by cisplatin resistance. Consistently, SKO cells were more responsive to mTOR inhibition by rapamycin and presented impairment of the feedback activation loop in Akt. Therefore, rapamycin was even more potent in inhibiting the clonogenic potential in SKO cells and sensitized to cisplatin treatment. Mechanistically, STAT3 partially coordinated the cisplatin resistance phenotype via the mTOR pathway in non-small cell lung cancer. Thus, our findings reveal important targets and highlight the significance of the crosstalk between STAT3 and mTOR signaling in cisplatin resistance. The synergic inhibition of STAT3 and mTOR potentially unveil a potential mechanism of synthetic lethality to be explored for human lung cancer treatment.
Collapse
Affiliation(s)
- Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Tharcísio Citrângulo Tortelli
- Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil; Laboratory of Signaling Mechanisms, School of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
| | - Luiz Guilherme Salvino Silva
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Daniela Campos Granato
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, Brazil
| | - Nathalie Fortes Pestana
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Luis Gustavo Saboia Ponte
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Guilherme Francisco Peruca
- Exercise Cell Biology Laboratory, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Bianca Alves Pauletti
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, Brazil
| | | | - Leandro Pereira de Moura
- Exercise Cell Biology Laboratory, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Rosângela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil
| | - Adriana Franco Paes Leme
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, SP, Brazil.
| |
Collapse
|
9
|
Ye X, Tang X, Zhao S, Ruan J, Wu M, Wang X, Li H, Zhong B. Mechanism of the growth and development of the posterior silk gland and silk secretion revealed by mutation of the fibroin light chain in silkworm. Int J Biol Macromol 2021; 188:375-384. [PMID: 34371049 DOI: 10.1016/j.ijbiomac.2021.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/12/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Silkworm, as a model organism, has very high economic value due to its silk secretion ability. Although a large number of studies have attempted to elucidate the mechanism of silk secretion, it remains unclear. In this study, the fibroin light chain (Fib-L) gene of silkworm was subjected to CRISPR/Cas9 editing, which yielded premature termination of translation at 135 aa. Compared with those of the wild type, the posterior silk glands (PSGs) of the homozygous mutants on the third day of the fifth instar showed obvious premature degeneration. Comparative transcriptome and proteomic analyses of the PSGs of wild-type individuals, heterozygous mutants and homozygous mutants were performed on the fourth day of the fifth instar. A GO enrichment analysis showed that the differentially expressed genes (DEGs) between homozygous mutants and wild-type individuals were enriched in cytoskeleton-related terms, and a KEGG enrichment analysis showed that the upregulated DEGs between homozygous mutants and wild-type individuals were enriched in the phagosome and apoptosis pathways. These results indicated that apoptosis was activated prematurely in the PSGs of homozygous mutants. Furthermore, autophagy and heat shock response were activated in the PSGs of homozygous mutants, as demonstrated by an analysis of the DEGs related to autophagy and heat shock. A comparative proteomic analysis further confirmed that autophagy, apoptosis and the heat shock response were activated in the PSGs of homozygous mutants, which led to premature degradation of the PSGs. These results provide insights for obtaining a more in-depth understanding of the mechanism of silk secretion in silkworms.
Collapse
Affiliation(s)
- Xiaogang Ye
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Xiaoli Tang
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Shuo Zhao
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Jinghua Ruan
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Meiyu Wu
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Xiaoxiao Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Huiping Li
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China
| | - Boxiong Zhong
- College of Animal Sciences, Zhejiang University, Hangzhou, PR China.
| |
Collapse
|
10
|
Enășescu DA, Moisescu MG, Imre M, Greabu M, Ripszky Totan A, Stanescu-Spinu I, Burcea M, Albu C, Miricescu D. Lutein Treatment Effects on the Redox Status and Metalloproteinase-9 (MMP-9) in Oral Cancer Squamous Cells-Are There Therapeutical Hopes? MATERIALS 2021; 14:ma14112968. [PMID: 34072756 PMCID: PMC8199462 DOI: 10.3390/ma14112968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/03/2023]
Abstract
Carotenoids loaded in nanoparticles should be regarded as a promising way to increase the availability in healthy cells and to induce apoptosis in cancer. Lutein is a carotenoid that, in contrast to beta-carotene, has no known toxicities. Oral cancer represents one of the most frequent types of cancer world-wide with an incidence rate of about 9% of all types of cancer. Almost 95% of all oral cancers are represented by squamous cell carcinomas (OSCC). The aim of this study was to review and analyse the effects of lutein and Poly(d,l-lactide-co-glycolide) (PLGA) Nps containing lutein (Lut Nps) on oxidative stress biomarkers (OXSR-1, FOXO-3, TAC) and collagen degradation biomarker-MMP-9, in human cells BICR10 of buccal mucosa squamous carcinoma. Lut Nps were prepared by the emulsion-solvent evaporation method. MMP, OXSR-1, TAC, FOXO-3 and MMP-9 were measured in tumour cell lysates by the ELISA technique. Our results have shown that in Lut 100 cells and Lut Nps the OXSR1 (p < 0.001, p < 0.001) and TAC (p < 0.001, p < 0.001) values were significantly higher than in control cells. The Lut 100 and Lut Nps FOXO-3 levels revealed no significant differences versus the control. MMP-9 levels were significantly reduced (p < 0.001) in the Lut Nps cells versus control cells. In our study conditions, lutein and lutein Nps did not trigger an oxidative stress by ROS induction. However, lutein Nps treatment seemed to have a positive effect, by downregulating the MMP-9 levels. Loaded in Nps, lutein could be regarded as a protective factor against local invasiveness, in whose molecular landscape MMPs, and especially MMP-9 are the main actors.
Collapse
Affiliation(s)
- Dan Alexandru Enășescu
- Department of Biochemistry, Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania; (D.A.E.); (M.G.); (I.S.-S.); (D.M.)
| | - Mihaela Georgeta Moisescu
- Department Biophysics and Cellular Biotechnology, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania;
- Excellence Centre for Research in Biophysics and Cellular Biotechnology, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania
| | - Marina Imre
- Department of Complete Denture, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania;
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania; (D.A.E.); (M.G.); (I.S.-S.); (D.M.)
| | - Alexandra Ripszky Totan
- Department of Biochemistry, Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania; (D.A.E.); (M.G.); (I.S.-S.); (D.M.)
- Correspondence: (A.R.T.); (C.A.)
| | - Iulia Stanescu-Spinu
- Department of Biochemistry, Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania; (D.A.E.); (M.G.); (I.S.-S.); (D.M.)
| | - Marian Burcea
- Department of Ophthalmology, Faculty of General Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroilor Sanitari Blvd., 050474 Bucharest, Romania;
| | - Crenguta Albu
- Department of Genetics, Faculty of General Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroilor Sanitari Blvd., 050474 Bucharest, Romania
- Correspondence: (A.R.T.); (C.A.)
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dental Medicine, University of Medicine and Pharmacy Carol Davila, 8 Eroii Sanitari Blvd., Sector 5, 050474 Bucharest, Romania; (D.A.E.); (M.G.); (I.S.-S.); (D.M.)
| |
Collapse
|
11
|
Oxidative Stress in Cancer Cell Metabolism. Antioxidants (Basel) 2021; 10:antiox10050642. [PMID: 33922139 PMCID: PMC8143540 DOI: 10.3390/antiox10050642] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are important in regulating normal cellular processes whereas deregulated ROS leads to the development of a diseased state in humans including cancers. Several studies have been found to be marked with increased ROS production which activates pro-tumorigenic signaling, enhances cell survival and proliferation and drives DNA damage and genetic instability. However, higher ROS levels have been found to promote anti-tumorigenic signaling by initiating oxidative stress-induced tumor cell death. Tumor cells develop a mechanism where they adjust to the high ROS by expressing elevated levels of antioxidant proteins to detoxify them while maintaining pro-tumorigenic signaling and resistance to apoptosis. Therefore, ROS manipulation can be a potential target for cancer therapies as cancer cells present an altered redox balance in comparison to their normal counterparts. In this review, we aim to provide an overview of the generation and sources of ROS within tumor cells, ROS-associated signaling pathways, their regulation by antioxidant defense systems, as well as the effect of elevated ROS production in tumor progression. It will provide an insight into how pro- and anti-tumorigenic ROS signaling pathways could be manipulated during the treatment of cancer.
Collapse
|
12
|
Zhang ZY, Yu XL, Cai MD, Liu YH, Liu JQ, Zhao SY, Li XX, Li YH. Relationship between bovine oocytes developmental competence and mRNA expression of apoptotic and mitochondrial genes following the change of vitrification temperatures and cryoprotectant concentrations. Cryobiology 2020; 97:110-122. [PMID: 33011172 DOI: 10.1016/j.cryobiol.2020.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/09/2023]
Abstract
The present study analyzed the relationship between bovine oocytes developmental competence and mRNA expression of apoptotic and mitochondrial genes following the change of vitrification temperatures (VTs) and cryoprotectant agent concentrations (CPAs). Cumulus oocyte complexes were randomly divided into five groups: control, vitrified in liquid nitrogen (LN; -196 °C) with 5.6 M CPAs (LN 5.6 M), LN with 6.6 M CPAs (LN 6.6 M), liquid helium (LHe; -269 °C) with 5.6 M CPAs (LHe 5.6 M), and LHe with 6.6 M CPAs (LHe 6.6 M). After vitrification and warming, oocytes of vitrified and control groups were subjected to in vitro maturation (IVM), in vitro fertilization and in vitro culture. The blastocyst rate in LHe 5.6 M group was the highest among the four vitrified groups (13.7% vs. 9.4%, 1.3%, and 8.4%; P < 0.05). The mRNA expression level of 8 apoptotic- and 12 mitochondria-related genes were detected through qRT-PCR after IVM. Lower VT (LHe, -269 °C) positively affected the mRNA expression levels of apoptotic genes (BAD, BID, BTK, TP53, and TP53I3) and mitochondrial genes (COX6B1, DERA, FIS1, NDUFA1, NDUFA4, PRDX2, SLC25A5, TFB1M, and UQCRB), and reduced oxidative stress from freezing. Decreased CPAs (5.6 M) positively affected mRNA expression levels of apoptotic genes (BAD, BCL2A1, BID, and CASP3) in LHe vitrification but negatively affected apoptotic genes (BAD, BAX, BID, BTK, and BCL2A1) in LN vitrification. In conclusion, decreased VTs and CPAs in LHe vitrification may increase the blastocyst rate by changing the mRNA expression levels of these apoptotic and mitochondrial genes for the vitrified oocytes.
Collapse
Affiliation(s)
- Zhi Yang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xue Li Yu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Meng Dan Cai
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Yi Heng Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Jia Qi Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Shi Yu Zhao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiao Xia Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| | - Ying Hua Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China; Henan Provincial Key Laboratory for Grass-Feeding Animal, Henan University of Science and Technology, Luoyang, 471023, China
| |
Collapse
|
13
|
Murray D, Mirzayans R. Nonlinearities in the cellular response to ionizing radiation and the role of p53 therein. Int J Radiat Biol 2020; 97:1088-1098. [PMID: 31986075 DOI: 10.1080/09553002.2020.1721602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 10/25/2022]
Abstract
Many aspects of the cellular response to agents such as ionizing radiation that cause genotoxic and/or oxidative stress exhibit a nonlinear relationship to the applied stress level. These include elements of the antioxidant response and of the damage-signaling pathways that determine cell fate decisions. The wild-type p53 protein, which is mutated in many cancers, coordinates these responses and is a key determinant of this nonlinearity. Indeed, p53 has been referred to as a 'cellular rheostat' that favors antioxidant/cytoprotective functions at low stress levels while switching to a pro-oxidant/cytotoxic role under high-stress conditions. For solid tumor-derived cell lines, moderate doses of radiation, typical of those used to generate clonogenic survival curves (i.e. ≤10 Gy), predominantly invoke a dose-dependent cytostatic response. For cancer cell lines with wild-type p53, cytostasis is primarily associated with features of senescence, whereas cancer cells with aberrant p53 primarily undergo endopolyploidization and enlargement. In line with a commentary by Meyn et al. [Int J Radiat Biol. 2009, 85:107-115] concluding that apoptosis is not the primary cause of radiation-induced loss of clonogenicity in solid tumor-derived cell lines, significant levels of apoptosis are typically seen only after higher doses (≥5 Gy) and this is almost all of the delayed (rather than primary) type. Nonlinearity of the oxidative/genotoxic stress response is already apparent in the early antioxidant events activated by transcription factors such as p53 and Nrf2 and the Ref1 transcription coactivator. These cytoprotective pathways serve to minimize damage to important cellular targets caused by reactive oxygen species (ROS) and other electrophiles. After high/supra-lethal levels of stress these inducible antioxidant pathways can be deactivated in a manner that would reinforce the establishment of the pro-oxidant state, resulting in elevated ROS levels and to cytostasis or apoptosis. Understanding the complex regulation of these damage-signaling pathways in relation to the stress levels is important for the optimal utilization of radiation therapy for cancer.
Collapse
Affiliation(s)
- David Murray
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
14
|
Selective inhibition of Aurora A and B kinases effectively induces cell cycle arrest in t(8;21) acute myeloid leukemia. Biomed Pharmacother 2019; 117:109113. [PMID: 31207577 DOI: 10.1016/j.biopha.2019.109113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/28/2019] [Accepted: 06/10/2019] [Indexed: 11/22/2022] Open
Abstract
The fusion gene AML1-ETO initially dysregulates various cell cycle molecules in t(8;21) acute myeloid leukemia. Aurora kinases have shown great promise in treating tumors. However, the efficacy of Aurora kinase (AURK) A and B inhibition in t(8;21) AML remains unclear. We found that AURK-A inhibitor Alisertib and AURK-B inhibitor Barasertib strongly inhibited the growth and proliferation of t(8;21) AML cells. The quantity and size of cell colonies were markedly decreased after a 14-d drug exposure. The cell cycle distribution was blocked at the G2/M phase in both dose- and time-dependent manner. The expression of p53 family and cdc2-p34 significantly changed as well. Notably, we found that t(8;21) AML cells are more sensitive to Aurora B inhibition. In each set of experiments, Barasertib took less time or a lower concentration to achieve similar efficacy. Taken together, our data highlighted the potential role of Aurora kinases as promising cell cycle targets for the treatment of t(8;21) AML and hereby provided a theoretical basis to guide relevant clinical trials.
Collapse
|
15
|
Duncan JR, Lieber MR, Adachi N, Wahl RL. Reply: Radiation Dose Does Matter: Mechanistic Insights into DNA Damage and Repair Support the Linear No-Threshold Model of Low-Dose Radiation Health Risks. J Nucl Med 2018; 59:1780-1781. [DOI: 10.2967/jnumed.118.218321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
16
|
Murray D, Mirzayans R, McBride WH. Defenses against Pro-oxidant Forces - Maintenance of Cellular and Genomic Integrity and Longevity. Radiat Res 2018; 190:331-349. [PMID: 30040046 PMCID: PMC6203329 DOI: 10.1667/rr15101.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There has been enormous recent progress in understanding how human cells respond to oxidative stress, such as that caused by exposure to ionizing radiation. We have witnessed a significant deciphering of the events that underlie how antioxidant responses counter pro-oxidant damage to key biological targets in all cellular compartments, including the genome and mitochondria. These cytoprotective responses include: 1. The basal cellular repertoire of antioxidant capabilities and its supporting cast of facilitator enzymes; and 2. The inducible phase of the antioxidant response, notably that mediated by the Nrf2 transcription factor. There has also been frenetic progress in defining how reactive electrophilic species swamp existing protective mechanisms to augment DNA damage, events that are embodied in the cellular "DNA-damage response", including cell cycle checkpoint activation and DNA repair, which occur on a time scale of hours to days, as well as the implementation of cellular responses such as apoptosis, autophagy, senescence and reprograming that extend the time period of damage sensing and response into weeks, months and years. It has become apparent that, in addition to the initial oxidative insult, cells typically undergo further waves of secondary reactive oxygen/nitrogen species generation, DNA damage and signaling and that these may reemerge long after the initial events have subsided, probably being driven, at least in part, by persisting DNA damage. These reactive oxygen/nitrogen species are an integral part of the pathological consequences of radiation exposure and may persist across multiple cell divisions. Because of the pervasive nature of oxidative stress, a cell will manifest different responses in different subcellular compartments and to different levels of stress injury. Aspects of these compartmentalized responses can involve the same proteins (such as ATM, p53 and p21) but in different functional guises, e.g., in cytoplasmic versus nuclear responses or in early- versus late-phase events. Many of these responses involve gene activation and new protein synthesis as well as a plethora of post-translational modifications of both basal and induced response proteins. It is these responses that we focus on in this review.
Collapse
Affiliation(s)
- David Murray
- Department of Oncology, Division of Experimental Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Canada
| | - Razmik Mirzayans
- Department of Oncology, Division of Experimental Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Canada
| | - William H. McBride
- Department of Radiation Oncology, University of California, Los Angeles (UCLA), Los Angeles, California
| |
Collapse
|
17
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Wang S, Zhang M, Liu P, Xie S, Cheng F, Wang L. Formation of pyrimidine-pyrimidine type DNA intrastrand cross-links: a theoretical verification. Phys Chem Chem Phys 2018; 19:28907-28916. [PMID: 29057416 DOI: 10.1039/c7cp06452g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pyrimidine-type radicals have been demonstrated to be able to attack their 3' or 5' neighboring purine nucleotides forming diverse DNA intrastrand cross-links, but whether or not these radicals can attack their surrounding pyrimidine nucleotides forming pyrimidine-pyrimidine type DNA intrastrand cross-links remains unclear. To resolve this question, probable additions of the uracil-5-methyl (˙UCH2) radical to the C5[double bond, length as m-dash]C6 double bond of its 3'/5' neighboring pyrimidine nucleotides in the four models, 5'-T(˙UCH2)-3', 5'-C(˙UCH2)-3', 5'-(˙UCH2)T-3', and 5'-(˙UCH2)C-3', are explored in the present work employing density functional theory (DFT) methods. The C6 site of its 5' neighboring thymidine is the preferred target for ˙UCH2 radical addition, while additions of the ˙UCH2 radical to the C6 and C5 sites of its 5' neighboring deoxycytidine are found to be competitive reactions. The ˙UCH2 radical can react with both the C6 and C5 sites of its 3' neighboring pyrimidine nucleotides, but the efficiencies of these reactions are predicted to be much lower than those of the corresponding addition reactions to its 5' neighboring pyrimidine nucleotides, indicating the existence of an obvious sequence effect. All the addition products could be finally transformed into closed-shell intrastrand cross-links, the molecular masses of which are found to be exactly the same as certain MS values determined in a recent study of an X-irradiated deoxygenated aqueous solution of calf thymus DNA. The present study thus not only definitely corroborates the fact that the reactive ˙UCH2 radical can attack its 3'/5' neighboring pyrimidine nucleotides forming several pyrimidine-pyrimidine type DNA intrastrand cross-links, but also provides a plausible explanation for the identities of these structurally unknown intrastrand cross-links.
Collapse
Affiliation(s)
- Shoushan Wang
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials, School of Environment and Civil Engineering, Dongguan University of Technology, Dongguan 523808, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
19
|
Wang S, Zhang M, Liu P, Xie S, Cheng F, Wang L. Mechanism studies of addition reactions between the pyrimidine type radicals and their 3′/5′ neighboring deoxyguanosines. RSC Adv 2018; 8:2777-2785. [PMID: 35541474 PMCID: PMC9077473 DOI: 10.1039/c7ra12713h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/06/2018] [Indexed: 11/21/2022] Open
Abstract
To clarify the biologically significant sequence effect existing in the formation of the pyrimidine-type radicals induced DNA intrastrand cross-links, addition mechanisms between the uridine-5-methyl (˙UCH2), 6-hydroxy-5,6-dihydrothymidine-5-yl (˙T6OH), and 6-hydroxy-5,6-dihydrocytidine-5-yl (˙C6OH) radicals and their 3′/5′ neighboring deoxyguanosines (dG) are explored in the present study employing the model 5′-G(˙UCH2)-3′, 5′-(˙UCH2)G-3′, 5′-G(˙T6OH)-3′, 5′-(˙T6OH)G-3′, 5′-G(˙C6OH)-3′, and 5′-(˙C6OH)G-3′ sequences. It is found that the 5′ G/C8 additions of the three radicals are all simple direct one-step reactions inducing only relatively small structural changes, while a conformational adjustment involving orientation transitions of both nucleobase moieties and twisting of the DNA backbone is indispensable for each 3′ G/C8 addition. Furthermore, markedly positive reaction free energy requirements are estimated for these conformational transformations making the 3′ G/C8 additions of the three radicals thermodynamically much more unfavorable than the corresponding 5′ G/C8 additions. Such essential conformational adjustments along the 3′ G/C8 addition paths that structurally greatly influence the local DNA structures and thermodynamically substantially reduce the addition efficiencies may be the reasons responsible for the differences in the formation yields and biological consequences of the pyrimidine-type radicals induced DNA intrastrand cross-link lesions. For each radical, the 5′ G/C8 addition is a simple direct one-step reaction, while a structurally significant and thermodynamically markedly unfavorable conformational adjustment is indispensable for the 3′ G/C8 addition.![]()
Collapse
Affiliation(s)
- Shoushan Wang
- School of Chemistry and Chemical Engineering
- South China University of Technology
- Guangzhou 510641
- People's Republic of China
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials
| | - Min Zhang
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials
- School of Environment and Civil Engineering
- Dongguan University of Technology
- Dongguan 523808
- People's Republic of China
| | - Peng Liu
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials
- School of Environment and Civil Engineering
- Dongguan University of Technology
- Dongguan 523808
- People's Republic of China
| | - Shilei Xie
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials
- School of Environment and Civil Engineering
- Dongguan University of Technology
- Dongguan 523808
- People's Republic of China
| | - Faliang Cheng
- Guangdong Engineering and Technology Research Center for Advanced Nanomaterials
- School of Environment and Civil Engineering
- Dongguan University of Technology
- Dongguan 523808
- People's Republic of China
| | - Lishi Wang
- School of Chemistry and Chemical Engineering
- South China University of Technology
- Guangzhou 510641
- People's Republic of China
| |
Collapse
|
20
|
Jin M, Park SJ, Kim SW, Kim HR, Hyun JW, Lee JH. PIG3 Regulates p53 Stability by Suppressing Its MDM2-Mediated Ubiquitination. Biomol Ther (Seoul) 2017; 25:396-403. [PMID: 28605833 PMCID: PMC5499618 DOI: 10.4062/biomolther.2017.086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 04/20/2017] [Accepted: 04/22/2017] [Indexed: 11/24/2022] Open
Abstract
Under normal, non-stressed conditions, intracellular p53 is continually ubiquitinated by MDM2 and targeted for degradation. However, in response to severe genotoxic stress, p53 protein levels are markedly increased and apoptotic cell death is triggered. Inhibiting the ubiquitination of p53 under conditions where DNA damage has occurred is therefore crucial for preventing the development of cancer, because if cells with severely damaged genomes are not removed from the population, uncontrolled growth can result. However, questions remain about the cellular mechanisms underlying the regulation of p53 stability. In this study, we show that p53-inducible gene 3 (PIG3), which is a transcriptional target of p53, regulates p53 stability. Overexpression of PIG3 stabilized both endogenous and transfected wild-type p53, whereas a knockdown of PIG3 lead to a reduction in both endogenous and UV-induced p53 levels in p53-proficient human cancer cells. Using both in vivo and in vitro ubiquitination assays, we found that PIG3 suppressed both ubiquitination- and MDM2-dependent proteasomal degradation of p53. Notably, we demonstrate that PIG3 interacts directly with MDM2 and promoted MDM2 ubiquitination. Moreover, elimination of endogenous PIG3 in p53-proficient HCT116 cells decreased p53 phosphorylation in response to UV irradiation. These results suggest an important role for PIG3 in regulating intracellular p53 levels through the inhibition of p53 ubiquitination.
Collapse
Affiliation(s)
- Min Jin
- Laboratory of Genomic Instability and Cancer therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| | - Seon-Joo Park
- Laboratory of Genomic Instability and Cancer therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.,Department of Premedical Sciences, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| | - Seok Won Kim
- Laboratory of Genomic Instability and Cancer therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.,Department of Neurosurgery, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| | - Hye Rim Kim
- Laboratory of Genomic Instability and Cancer therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung-Hee Lee
- Laboratory of Genomic Instability and Cancer therapeutics, Cancer Mutation Research Center, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| |
Collapse
|
21
|
Wu G, Wang N, Luo Y, Zhang Y, Wang P, Zhu Z, Gao Y, Du Z, Yang B. Metabolic perturbation of epigenome by inhibiting S-adenosylhomocysteine hydrolase elicits senescence through DNA damage response in hepatoma cells. Tumour Biol 2017; 39:1010428317699117. [PMID: 28459194 DOI: 10.1177/1010428317699117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence is a key physiological barrier against tumor and represents an option for therapeutic intervention. One pivotal intracellular stimulus causing senescence is DNA damage response, while the senescence-associated heterochromatin in cancer limits the strength of the DNA damage response to endogenous genotoxic stress or DNA-damaging agents. Therefore, targeting the maintenance of compacted chromatin in cancer cells represents an optional intervention to improve the therapeutic efficacy in cancer treatment. Given a crosstalk between methionine cycle and histone methylation, we hypothesize that pharmacologically disrupting methylation potential, defined as the ratio of cellular S-adenosylmethionine to S-adenosylhomocysteine, could affect the chromatin structures in cancer cells and thus enhance their sensitivity to DNA damage response signaling. Our results showed that 3-deazaneplanocin A, a chemical inhibitor of S-adenosylhomocysteine hydrolase, elicited a typical cellular senescence in hepatoma cells. Therapy-induced senescence by 3-deazaneplanocin A was mediated through p53-p21 pathway and triggered by enhanced ataxia-telangiectasia mutated activation related to chromatin changes. In conclusion, our study demonstrated that metabolic perturbation of chromatin status in oncogene-activated cancers could be an optional intervention to sensitize DNA damage response signaling.
Collapse
Affiliation(s)
- Guozhen Wu
- 1 Clinical School, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 2 Department of Hepatobiliary Surgery, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Ning Wang
- 3 Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Ying Luo
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Yanyan Zhang
- 1 Clinical School, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Peng Wang
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Zhengyan Zhu
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Yingtang Gao
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Zhi Du
- 2 Department of Hepatobiliary Surgery, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| | - Bin Yang
- 4 Key Laboratory of Artificial Cells, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
- 5 Institute of Hepatobiliary Disease, The Third Central Hospital of Tianjin, Tianjin Medical University, Tianjin, China
| |
Collapse
|
22
|
Chen H, Li Y, Long Y, Tang E, Wang R, Huang K, Xie C, Chen G. Increased p16 and p53 protein expression predicts poor prognosis in mucosal melanoma. Oncotarget 2017; 8:53226-53233. [PMID: 28881806 PMCID: PMC5581105 DOI: 10.18632/oncotarget.18367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/10/2017] [Indexed: 12/14/2022] Open
Abstract
Primary mucosal melanoma (MM) is a rare, and aggressive, neoplasm with a poor prognosis. To date, few prognostic markers of MM have been well-defined. The aim of this study is to clarify the prognostic value of p53 and p16 proteins in predicting the clinical outcome of Chinese patients with MM. A total of 59 MM samples were contained from biopsy specimens, and, expressions of p53 and p16 proteins were assessed by immunohistochemistry. Cox regression analysis was performed to investigate the association of these proteins with the overall survival of MM patients. Increased p16 expression was significantly associated with reduced survival at three years (P=0.039). Increased p53 expression correlates with reduced one-year (P=0.025), and, two-year survival (P=0.037). Increased p53 and p16 protein expression may be helpful prognostic indicators for the management of these patients.
Collapse
Affiliation(s)
- Hanbin Chen
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangyang Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yin Long
- Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Yangpu, Shanghai, China
| | - Erjiang Tang
- Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Yangpu, Shanghai, China
| | - Rongrong Wang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kate Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congying Xie
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guorong Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
Gào X, Schöttker B. Reduction-oxidation pathways involved in cancer development: a systematic review of literature reviews. Oncotarget 2017; 8:51888-51906. [PMID: 28881698 PMCID: PMC5584299 DOI: 10.18632/oncotarget.17128] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/03/2017] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress results from an imbalance of the reactive oxygen species/reactive nitrogen species (ROS/RNS) production and the oxidants defense system. Extensive research during the last decades has revealed that oxidative stress can mediate cancer initiation and development by leading not only to molecular damage but also to a disruption of reduction-oxidation (redox) signaling. In order to provide a global overview of the redox signaling pathways, which play a role in cancer formation, we conducted a systematic literature search in PubMed and ISI Web of Science and identified 185 relevant reviews published in the last 10 years. The 20 most frequently described pathways were selected to be presented in this systematic review and could be categorized into 3 groups: Intracellular ROS/RNS generating organelles and enzymes, signal transduction cascades kinases/phosphatases and transcription factors. Intracellular ROS/RNS generation organelles are mitochondria, endoplasmic reticulum and peroxisomes. Enzymes, including NOX, COX, LOX and NOS, are the most prominent enzymes generating ROS/RNS. ROS/RNS act as redox messengers of transmembrane receptors and trigger the activation or inhibition of signal transduction kinases/phosphatases, such as the family members of protein tyrosine kinases and protein tyrosine phosphatases. Furthermore, these reactions activate downstream signaling pathways including protein kinase of the MAPK cascade, PI3K and PKC. The kinases and phosphatases regulate the phosphorylation status of transcription factors including APE1/Ref-1, HIF-1α, AP-1, Nrf2, NF-κB, p53, FOXO, STAT, and β-catenin. Finally, we briefly discuss cancer prevention and treatment opportunities, which address redox pathways and further research needs.
Collapse
Affiliation(s)
- Xīn Gào
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,Network Aging Research, University of Heidelberg, Heidelberg, Germany.,Institute of Health Care and Social Sciences, FOM University, Essen, Germany
| |
Collapse
|
24
|
Quan J, Li Y, Jin M, Chen D, Yin X, Jin M. Suppression of p53-inducible gene 3 is significant for glioblastoma progression and predicts poor patient prognosis. Tumour Biol 2017; 39:1010428317694572. [PMID: 28351326 DOI: 10.1177/1010428317694572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma is the most malignant and invasive brain tumor with extremely poor prognosis. p53-inducible gene 3, a downstream molecule of the tumor suppressor p53, has been found involved in apoptosis and oxidative stress response. However, the functions of p53-inducible gene 3(PIG3) in cancer are far from clear including glioblastoma. In this study, we found that p53-inducible gene 3 expression was suppressed in glioblastoma tissues compared with normal tissues. And the expression of p53-inducible gene 3 was significantly associated with the World Health Organization grade. Patients with high p53-inducible gene 3 expression have a significantly longer median survival time (15 months) than those with low p53-inducible gene 3 expression (8 months). According to Cox regression analysis, p53-inducible gene 3 was an independent prognostic factor with multivariate hazard ratio of 0.578 (95% confidence interval, 0.352-0.947; p = 0.030) for overall survival. Additionally, gain and loss of function experiments showed that knockdown of p53-inducible gene 3 significantly increased the proliferation and invasion ability of glioblastoma cells while overexpression of p53-inducible gene 3 inhibited the proliferation and invasion ability. The results of in vivo glioblastoma models further confirmed that p53-inducible gene 3 suppression promoted glioblastoma progression. Altogether, our data suggest that high expression of p53-inducible gene 3 is significant for glioblastoma inhibition and p53-inducible gene 3 independently indicates good prognosis in patients, which might be a novel prognostic biomarker or potential therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Jishu Quan
- 1 Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, China
| | - Yong Li
- 2 Department of Pediatrics, Yanbian Maternity and Child Care Center, Yanji, China
| | - Meihua Jin
- 1 Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, China
| | - Dunfu Chen
- 1 Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, China
| | - Xuezhe Yin
- 3 Yanbian University Hospital, Yanji, China
| | - Ming Jin
- 1 Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, China
| |
Collapse
|
25
|
Li M, Li S, Liu B, Gu MM, Zou S, Xiao BB, Yu L, Ding WQ, Zhou PK, Zhou J, Shang ZF. PIG3 promotes NSCLC cell mitotic progression and is associated with poor prognosis of NSCLC patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:39. [PMID: 28259183 PMCID: PMC5336678 DOI: 10.1186/s13046-017-0508-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/21/2017] [Indexed: 02/07/2023]
Abstract
Background Non-small cell lung cancer (NSCLC) is the most commonly diagnosed type of lung cancer that is associated with poor prognosis. In this study we explored the potential role of p53-induced gene 3 (PIG3) in the progression of NSCLC. Methods Immunohistochemistry was used to determine the expression levels of PIG3 in 201 NSCLC patients. We performed in vitro studies and silenced endogenous PIG3 by using specific siRNAs that specific target PIG3. Immunofluorescent staining was performed to determine the effect of PIG3 on mitotic progression in NSCLC cells. The growth rates of microtubules were determined by microtubule nucleation analysis. Cell proliferation and chemosensitivity were analyzed by CCK8 assays. Annexin V staining and β-galactosidase activity analysis were used to evaluate PIG3 deficiency-related apoptosis and senescence, respectively. Results PIG3 expression levels negatively correlated with overall survival and disease-free survival of NSCLC patients. Knock down of PIG3 resulted in repressed proliferation of NSCLC cells and increased aberrant mitosis, which included misaligning and lagging chromosomes, and bi- or multi-nucleated giant cells. In addition, PIG3 contributed to mitotic spindle assembly by promoting microtubule growth. Furthermore, loss of PIG3 sensitized NSCLC cells to docetaxel by enhancing docetaxel-induced apoptosis and senescence. Conclusions Our results indicate that PIG3 promotes NSCLC progression and therefore suggest that PIG3 may be a potential prognostic biomarker and novel therapeutic target for the treatment of NSCLC. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0508-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ming Li
- 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, People's Republic of China
| | - Shanhu Li
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China
| | - Biao Liu
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, 215001, People's Republic of China
| | - Meng-Meng Gu
- 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, People's Republic of China
| | - Shitao Zou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, 215001, People's Republic of 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, People's Republic of China
| | - Lan Yu
- Department of Radiation Oncology, Simmons Comprehensive Cancer Center at UT Southwestern Medical Center, Dallas, 75390, TX, USA
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - Ping-Kun Zhou
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, 215001, People's Republic of 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, People's Republic of China. .,Department of Radiation Oncology, Simmons Comprehensive Cancer Center at UT Southwestern Medical Center, Dallas, 75390, TX, USA.
| |
Collapse
|
26
|
Tiveron AP, Rosalen PL, Franchin M, Lacerda RCC, Bueno-Silva B, Benso B, Denny C, Ikegaki M, de Alencar SM. Chemical Characterization and Antioxidant, Antimicrobial, and Anti-Inflammatory Activities of South Brazilian Organic Propolis. PLoS One 2016; 11:e0165588. [PMID: 27802316 PMCID: PMC5089781 DOI: 10.1371/journal.pone.0165588] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/16/2016] [Indexed: 11/28/2022] Open
Abstract
South Brazilian organic propolis (OP), which has never been studied before, was assessed and its chemical composition, scavenging potential of reactive oxygen species, antimicrobial and anti-inflammatory activities are herein presented. Based on the chemical profile obtained using HPLC, OP was grouped into seven variants (OP1–OP7) and all of them exhibited high scavenging activity, mainly against superoxide and hypochlorous acid species. OP1, OP2, and OP3 had the smallest minimal inhibitory concentration (MIC) against Gram-positive bacteria Streptococcus mutans, Streptococcus oralis, and Streptococcus aureus (12.5–100 μg/mL). OP1, OP2, OP3, and OP4 were more effective against Pseudomonas aeruginosa (Gram-negative), with MIC values ranging from 100 to 200 μg/mL. OP6 showed anti-inflammatory activity by decreasing NF-kB activation and TNF-α release in RAW 264.7 macrophages, and expressing the NF-κB-luciferase reporter stable gene. Therefore, south Brazilian OP can be considered an excellent source of bioactive compounds with great potential of application in the pharmaceutical and food industry.
Collapse
Affiliation(s)
- Ana Paula Tiveron
- Department of Agri-Food Industry, Food and Nutrition, “Luiz de Queiroz” College of Agriculture, University of São Paulo (USP), Avenida Pádua Dias, 11, 13418-900, Piracicaba, SP, Brazil
| | - Pedro Luiz Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av. Limeira, 901, CP 52, 13414-903, Piracicaba, SP, Brazil
| | - Marcelo Franchin
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av. Limeira, 901, CP 52, 13414-903, Piracicaba, SP, Brazil
| | - Risia Cristina Coelho Lacerda
- Department of Agri-Food Industry, Food and Nutrition, “Luiz de Queiroz” College of Agriculture, University of São Paulo (USP), Avenida Pádua Dias, 11, 13418-900, Piracicaba, SP, Brazil
| | - Bruno Bueno-Silva
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av. Limeira, 901, CP 52, 13414-903, Piracicaba, SP, Brazil
| | - Bruna Benso
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av. Limeira, 901, CP 52, 13414-903, Piracicaba, SP, Brazil
| | - Carina Denny
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av. Limeira, 901, CP 52, 13414-903, Piracicaba, SP, Brazil
| | - Masaharu Ikegaki
- School of Pharmaceutical Sciences, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 714, Centro, 37130-000, Alfenas, MG, Brazil
| | - Severino Matias de Alencar
- Department of Agri-Food Industry, Food and Nutrition, “Luiz de Queiroz” College of Agriculture, University of São Paulo (USP), Avenida Pádua Dias, 11, 13418-900, Piracicaba, SP, Brazil
- * E-mail:
| |
Collapse
|
27
|
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: 3.3] [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
|
28
|
Di Mascio P, Martinez GR, Miyamoto S, Ronsein GE, Medeiros MH, Cadet J. Singlet molecular oxygen: Düsseldorf – São Paulo, the Brazilian connection. Arch Biochem Biophys 2016; 595:161-75. [DOI: 10.1016/j.abb.2015.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/28/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022]
|
29
|
New Therapeutic Concept of NAD Redox Balance for Cisplatin Nephrotoxicity. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4048390. [PMID: 26881219 PMCID: PMC4736397 DOI: 10.1155/2016/4048390] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/09/2015] [Indexed: 12/14/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent for the treatment of various tumors. In addition to its antitumor activity, cisplatin affects normal cells and may induce adverse effects such as ototoxicity, nephrotoxicity, and peripheral neuropathy. Various mechanisms such as DNA adduct formation, mitochondrial dysfunction, oxidative stress, and inflammatory responses are closely associated with cisplatin-induced nephrotoxicity; however, the precise mechanism remains unclear. The cofactor nicotinamide adenine dinucleotide (NAD+) has emerged as a key regulator of cellular energy metabolism and homeostasis. Recent studies have demonstrated associations between disturbance in intracellular NAD+ levels and clinical progression of various diseases through the production of reactive oxygen species and inflammation. Furthermore, we demonstrated that reduction of the intracellular NAD+/NADH ratio is critically involved in cisplatin-induced kidney damage through inflammation and oxidative stress and that increase of the cellular NAD+/NADH ratio suppresses cisplatin-induced kidney damage by modulation of potential damage mediators such as oxidative stress and inflammatory responses. In this review, we describe the role of NAD+ metabolism in cisplatin-induced nephrotoxicity and discuss a potential strategy for the prevention or treatment of cisplatin-induced adverse effects with a particular focus on NAD+-dependent cellular pathways.
Collapse
|
30
|
Herraiz C, Calvo F, Pandya P, Cantelli G, Rodriguez-Hernandez I, Orgaz JL, Kang N, Chu T, Sahai E, Sanz-Moreno V. Reactivation of p53 by a Cytoskeletal Sensor to Control the Balance Between DNA Damage and Tumor Dissemination. J Natl Cancer Inst 2016; 108:djv289. [PMID: 26464464 PMCID: PMC4712681 DOI: 10.1093/jnci/djv289] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 04/22/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Abnormal cell migration and invasion underlie metastasis, and actomyosin contractility is a key regulator of tumor invasion. The links between cancer migratory behavior and DNA damage are poorly understood. METHODS Using 3D collagen systems to recapitulate melanoma extracellular matrix, we analyzed the relationship between the actomyosin cytoskeleton of migrating cells and DNA damage. We used multiple melanoma cell lines and microarray analysis to study changes in gene expression and in vivo intravital imaging (n = 7 mice per condition) to understand how DNA damage impacts invasive behavior. We used Protein Tissue Microarrays (n = 164 melanomas) and patient databases (n = 354 melanoma samples) to investigate the associations between markers of DNA damage and actomyosin cytoskeletal features. Data were analyzed with Student's and multiple t tests, Mann-Whitney's test, one-way analysis of variance, and Pearson correlation. All statistical tests were two-sided. RESULTS Melanoma cells with low levels of Rho-ROCK-driven actomyosin are subjected to oxidative stress-dependent DNA damage and ATM-mediated p53 protein stabilization. This results in a specific transcriptional signature enriched in DNA damage/oxidative stress responsive genes, including Tumor Protein p53 Inducible Protein 3 (TP53I3 or PIG3). PIG3, which functions in DNA damage repair, uses an unexpected catalytic mechanism to suppress Rho-ROCK activity and impair tumor invasion in vivo. This regulation was suppressed by antioxidants. Furthermore, PIG3 levels decreased while ROCK1/2 levels increased in human metastatic melanomas (ROCK1 vs PIG3; r = -0.2261, P < .0001; ROCK2 vs PIG3: r = -0.1381, P = .0093). CONCLUSIONS The results suggest using Rho-kinase inhibitors to reactivate the p53-PIG3 axis as a novel therapeutic strategy; we suggest that the use of antioxidants in melanoma should be very carefully evaluated.
Collapse
Affiliation(s)
- Cecilia Herraiz
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Fernando Calvo
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Pahini Pandya
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Gaia Cantelli
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Irene Rodriguez-Hernandez
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Jose L Orgaz
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - NaRa Kang
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Tinghine Chu
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Erik Sahai
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH)
| | - Victoria Sanz-Moreno
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK (CH, PP, GC, IRH, JLO, NK, TC, VSM); Tumor Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK (FC, ES).Current affiliations: Tumor Microenvironment Team, Institute of Cancer Research, Chester Beatty Laboratories, London, UK (FC); Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia and IMIB-Arrixaca, Murcia, Spain (CH).
| |
Collapse
|
31
|
Amino-terminal p53 mutations lead to expression of apoptosis proficient p47 and prognosticate better survival, but predispose to tumorigenesis. Proc Natl Acad Sci U S A 2015; 112:E6349-58. [PMID: 26578795 DOI: 10.1073/pnas.1510043112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Whereas most mutations in p53 occur in the DNA-binding domain and lead to its functional inactivation, their relevance in the amino-terminal transactivation domain is unclear. We show here that amino-terminal p53 (ATp53) mutations often result in the abrogation of full-length p53 expression, but concomitantly lead to the expression of the amino-terminally truncated p47 isoform. Using genetically modified cancer cells that only express p47, we demonstrate it to be up-regulated in response to various stimuli, and to contribute to cell death, through its ability to selectively activate a group of apoptotic target genes. Target gene selectivity is influenced by K382 acetylation, which depends on the amino terminus, and is required for recruitment of selective cofactors. Consistently, cancers capable of expressing p47 had a better overall survival. Nonetheless, retention of the apoptotic function appears insufficient for tumor suppression, because these mutations are also found in the germ line and lead to Li-Fraumeni syndrome. These data from ATp53 mutations collectively demonstrate that p53's apoptosis proficiency is dispensable for tumor suppression, but could prognosticate better survival.
Collapse
|
32
|
Xu J, Cai J, Jin X, Yang J, Shen Q, Ding X, Liang Y. PIG3 plays an oncogenic role in papillary thyroid cancer by activating the PI3K/AKT/PTEN pathway. Oncol Rep 2015; 34:1424-30. [PMID: 26133772 DOI: 10.3892/or.2015.4096] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/18/2015] [Indexed: 12/15/2022] Open
Abstract
The p53-inducible gene 3 (PIG3 or TP53I3) is a downstream gene of p53, which can be involved in the process of apoptosis induced by p53 via the production of reactive oxygen species (ROS). However, the functional significance of PIG3 in cancer remains to be determined. This aim of this study was to examine the mRNA and protein expression of PIG3 in papillary thyroid carcinoma (PTC) and normal thyroid tissues, assess the relationship between PIG3 expression and clinicopathological parameters in PTC and examine its role in the proliferation of PTC cell lines. The results showed that PIG3 was aberrantly overexpressed in the majority of specimens of PTC while the expression of p53 was lower in PTC compared with normal thyroid tissues. Anti-PIG3 immuno-reactivity positively correlated with TNM grade. In the PTC cell lines, PIG3 silencing using small interfering RNA (siRNAs) impaired their ability of proliferation and decreased the activity of the PI3K/AKT/PTEN pathway. The results suggested that PIG3 plays an oncogenic role in PTC via the regulation of the PI3K/AKT/PTEN pathway and support the exploration of PIG3 as a novel biomarker for patients with PTC.
Collapse
Affiliation(s)
- Jinye Xu
- Department of Oncologic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Junbo Cai
- Department of Oncologic Surgery, The Affiliated Municipal Hospital, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xiaoyan Jin
- Department of Oncologic Surgery, The Affiliated Municipal Hospital, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jingjin Yang
- Department of Clinical Laboratory, Medical School, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Qinyan Shen
- Department of Oncologic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaofei Ding
- Department of Clinical Laboratory, Medical School, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Yong Liang
- Department of Oncologic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
33
|
Kim HJ, Oh GS, Shen A, Lee SB, Khadka D, Pandit A, Shim H, Yang SH, Cho EY, Song J, Kwak TH, Choe SK, Park R, So HS. Nicotinamide adenine dinucleotide: An essential factor in preserving hearing in cisplatin-induced ototoxicity. Hear Res 2015; 326:30-9. [PMID: 25891352 DOI: 10.1016/j.heares.2015.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/07/2015] [Indexed: 12/20/2022]
Abstract
Ototoxicity is an important issue in patients receiving cisplatin chemotherapy. Numerous studies have demonstrated that several mechanisms, including oxidative stress, DNA damage, and inflammatory responses, are closely associated with cisplatin-induced ototoxicity. Although much attention has been directed at identifying ways to protect the inner ear from cisplatin-induced damage, the precise underlying mechanisms have not yet been elucidated. The cofactor nicotinamide adenine dinucleotide (NAD(+)) has emerged as an important regulator of cellular energy metabolism and homeostasis. NAD(+) acts as a cofactor for various enzymes including sirtuins (SIRTs) and poly(ADP-ribose) polymerases (PARPs), and therefore, maintaining adequate NAD(+) levels has therapeutic benefits because of its effect on NAD(+)-dependent enzymes. Recent studies demonstrated that disturbance in intracellular NAD(+) levels is critically involved in cisplatin-induced cochlear damage associated with oxidative stress, DNA damage, and inflammatory responses. In this review, we describe the importance of NAD(+) in cisplatin-induced ototoxicity and discuss potential strategies for the prevention or treatment of cisplatin-induced ototoxicity with a particular focus on NAD(+)-dependent cellular pathways.
Collapse
Affiliation(s)
- Hyung-Jin Kim
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Gi-Su Oh
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Su-Bin Lee
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Dipendra Khadka
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Hyeok Shim
- Department of Internal Medicine, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Sei-Hoon Yang
- Department of Internal Medicine, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Eun-Young Cho
- Department of Internal Medicine, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Jeho Song
- Department of Sports Industry and Welfare, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Tae Hwan Kwak
- PAEAN Biotechnology, 160 Techno-2 Street, Yuseong-gu, Daejeon, 305-500, Republic of Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Raekil Park
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation & Department of Microbiology, School of Medicine, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea.
| |
Collapse
|
34
|
Gerlei KZ, Yarligan Uysal S. A comparison of l- and d-Asp and Asn α-radicals a case study for atropisomerism. COMPUT THEOR CHEM 2014. [DOI: 10.1016/j.comptc.2014.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Havaki S, Kotsinas A, Chronopoulos E, Kletsas D, Georgakilas A, Gorgoulis VG. The role of oxidative DNA damage in radiation induced bystander effect. Cancer Lett 2014; 356:43-51. [PMID: 24530228 DOI: 10.1016/j.canlet.2014.01.023] [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: 08/15/2013] [Revised: 12/08/2013] [Accepted: 01/24/2014] [Indexed: 02/06/2023]
Abstract
Ionizing radiation (IR) has been described as a double-edged sword, since it is used for diagnostic and therapeutic medical applications, and at the same time it is a well known human mutagen and carcinogen, causing wide-ranging chromosomal aberrations. It is nowadays accepted that the detrimental effects of IR are not restricted only in the irradiated cells, but also to non-irradiated bystander or even distant cells manifesting various biological effects. This review presents the role of oxidative stress in the induction of bystander effects referring to the types of the implicated oxidative DNA lesions, the contributing intercellular and intracellular stress mediators, the way they are transmitted from irradiated to bystander cells and finally, the complex role of the bystander effect in the therapeutic efficacy of radiation treatment of cancer.
Collapse
Affiliation(s)
- Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | | | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Athens, Greece
| | - Alexandros Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece; Biomedical Research Foundation, Academy of Athens, Athens, Greece; Faculty Institute for Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, M13 9WL, UK.
| |
Collapse
|
36
|
Kalko SG, Paco S, Jou C, Rodríguez MA, Meznaric M, Rogac M, Jekovec-Vrhovsek M, Sciacco M, Moggio M, Fagiolari G, De Paepe B, De Meirleir L, Ferrer I, Roig-Quilis M, Munell F, Montoya J, López-Gallardo E, Ruiz-Pesini E, Artuch R, Montero R, Torner F, Nascimento A, Ortez C, Colomer J, Jimenez-Mallebrera C. Transcriptomic profiling of TK2 deficient human skeletal muscle suggests a role for the p53 signalling pathway and identifies growth and differentiation factor-15 as a potential novel biomarker for mitochondrial myopathies. BMC Genomics 2014; 15:91. [PMID: 24484525 PMCID: PMC3937154 DOI: 10.1186/1471-2164-15-91] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 01/28/2014] [Indexed: 02/08/2023] Open
Abstract
Background Mutations in the gene encoding thymidine kinase 2 (TK2) result in the myopathic form of mitochondrial DNA depletion syndrome which is a mitochondrial encephalomyopathy presenting in children. In order to unveil some of the mechanisms involved in this pathology and to identify potential biomarkers and therapeutic targets we have investigated the gene expression profile of human skeletal muscle deficient for TK2 using cDNA microarrays. Results We have analysed the whole transcriptome of skeletal muscle from patients with TK2 mutations and compared it to normal muscle and to muscle from patients with other mitochondrial myopathies. We have identified a set of over 700 genes which are differentially expressed in TK2 deficient muscle. Bioinformatics analysis reveals important changes in muscle metabolism, in particular, in glucose and glycogen utilisation, and activation of the starvation response which affects aminoacid and lipid metabolism. We have identified those transcriptional regulators which are likely to be responsible for the observed changes in gene expression. Conclusion Our data point towards the tumor suppressor p53 as the regulator at the centre of a network of genes which are responsible for a coordinated response to TK2 mutations which involves inflammation, activation of muscle cell death by apoptosis and induction of growth and differentiation factor 15 (GDF-15) in muscle and serum. We propose that GDF-15 may represent a potential novel biomarker for mitochondrial dysfunction although further studies are required.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Cecilia Jimenez-Mallebrera
- Neuromuscular Unit, Neurology Department, Fundación Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
37
|
Guan X, Liu Z, Wang L, Johnson DG, Wei Q. Identification of prohibitin and prohibiton as novel factors binding to the p53 induced gene 3 (PIG3) promoter (TGYCC)(15) motif. Biochem Biophys Res Commun 2014; 443:1239-44. [PMID: 24388982 DOI: 10.1016/j.bbrc.2013.12.124] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/23/2013] [Indexed: 01/30/2023]
Abstract
The promoter of p53 induced gene 3 (PIG3) contains a variable number of tandem repeats (VNTRs) of pentanucleotides (TGYCC)n that is known as a p53 binding site. In this study, we investigated whether other potential molecules could bind to this PIG3 promoter (TGYCC)n motif. Ligand-chromatography combined with liquid chromatography-tandem mass spectrometry analyses indicated direct interactions of prohibitin and/or prohibiton with the (TGYCC)15 motif, which was confirmed by electrophoretic mobility shift assay and super-gel shift analysis with anti-prohibitin and anti-prohibiton antibodies. Using the chromatin immunopercipipation assay, we further demonstrated that prohibitin and prohibiton associated with the (TGYCC)15 motif in vivo regardless of the p53 status and apoptotic stress. We also found that prohibitin and prohibiton up-regulated PIG3 transcription independent of p53, although p53 obviously enhanced this process, and that the knock-down of prohibitin and prohibiton inhibited camptothecin-induced apoptosis. Taken together, our findings suggest that prohibitin and prohibiton contribute to PIG3-mediated apoptosis by binding to the PIG3 promoter (TGYCC)15 motif.
Collapse
Affiliation(s)
- Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Zhensheng Liu
- Duke Cancer Institute, Duke University School of Medicine, USA
| | - Luo Wang
- Department of Internal Medicine, Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan Medical School, USA
| | - David G Johnson
- Department of Molecular Carcinogenesis, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University School of Medicine, USA.
| |
Collapse
|
38
|
Yu B, Li X, Zheng W, Feng Y, Wong YS, Chen T. pH-responsive cancer-targeted selenium nanoparticles: a transformable drug carrier with enhanced theranostic effects. J Mater Chem B 2014; 2:5409-5418. [DOI: 10.1039/c4tb00399c] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A cancer-targeted and structure-transformable drug delivery system has been constructed, which displays enhanced anticancer efficacy and exhibits the characteristics of shape transformation and pH-controlled drug release under acidifying cell organelles.
Collapse
Affiliation(s)
- Bo Yu
- Department of Chemistry
- Jinan University
- Guangzhou 510632, China
| | - Xiaoling Li
- Department of Chemistry
- Jinan University
- Guangzhou 510632, China
| | - Wenjie Zheng
- Department of Chemistry
- Jinan University
- Guangzhou 510632, China
| | - Yanxian Feng
- Department of Chemistry
- Jinan University
- Guangzhou 510632, China
| | - Yum-Shing Wong
- School of Life Science and State Key Laboratory for Agrobiotechnology
- The Chinese University of Hong Kong
- Hong Kong S.A.R, China
| | - Tianfeng Chen
- Department of Chemistry
- Jinan University
- Guangzhou 510632, China
| |
Collapse
|
39
|
Gerlei KZ, Jákli I, Szőri M, Jensen SJK, Viskolcz B, Csizmadia IG, Perczel A. Atropisomerism of the Asn α radicals revealed by Ramachandran surface topology. J Phys Chem B 2013; 117:12402-9. [PMID: 24015919 DOI: 10.1021/jp4070906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
C radicals are typically trigonal planar and thus achiral, regardless of whether they originate from a chiral or an achiral C-atom (e.g., C-H + (•)OH → C• + H2O). Oxidative stress could initiate radical formation in proteins when, for example, the H-atom is abstracted from the Cα-carbon of an amino acid residue. Electronic structure calculations show that such a radical remains achiral when formed from the achiral Gly, or the chiral but small Ala residues. However, when longer side-chain containing proteogenic amino acid residues are studied (e.g., Asn), they provide radicals of axis chirality, which in turn leads to atropisomerism observed for the first time for peptides. The two enantiomeric extended backbone structures, •βL and •βD, interconvert via a pair of enantiotopic reaction paths, monitored on a 4D Ramachandran surface, with two distinct transition states of very different Gibbs-free energies: 37.4 and 67.7 kJ/mol, respectively. This discovery requires the reassessment of our understanding on radical formation and their conformational and stereochemical behavior. Furthermore, the atropisomerism of proteogenic amino acid residues should affect our understanding on radicals in biological systems and, thus, reframes the role of the D-residues as markers of molecular aging.
Collapse
Affiliation(s)
- Klára Z Gerlei
- Department of Chemical Informatics, Faculty of Education, University of Szeged , 6726 Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
40
|
Gao S, Hsieh CL, Bhansali M, Kannan A, Shemshedini L. A peptide against soluble guanylyl cyclase α1: a new approach to treating prostate cancer. PLoS One 2013; 8:e64189. [PMID: 23724033 PMCID: PMC3664642 DOI: 10.1371/journal.pone.0064189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/13/2013] [Indexed: 11/26/2022] Open
Abstract
Among the many identified androgen-regulated genes, sGCα1 (soluble guanylyl cyclase α1) appears to play a pivotal role in mediating the pro-cancer effects of androgens and androgen receptor. The classical role for sGCα1 is to heterodimerize with the sGCβ1 subunit, forming sGC, the enzyme that mediates nitric oxide signaling by catalyzing the synthesis of cyclic guanosine monophosphate. Our published data show that sGCα1 can drive prostate cancer cell proliferation independent of hormone and provide cancer cells a pro-survival function, via a novel mechanism for p53 inhibition, both of which are independent of sGCβ1, NO, and cGMP. All of these properties make sGCα1 an important novel target for prostate cancer therapy. Thus, peptides were designed targeting sGCα1 with the aim of disrupting this protein’s pro-cancer activities. One peptide (A-8R) was determined to be strongly cytotoxic to prostate cancer cells, rapidly inducing apoptosis. Cytotoxicity was observed in both hormone-dependent and, significantly, hormone-refractory prostate cancer cells, opening the possibility that this peptide can be used to treat the usually lethal castration-resistant prostate cancer. In mouse xenograft studies, Peptide A-8R was able to stop tumor growth of not only hormone-dependent cells, but most importantly from hormone-independent cells. In addition, the mechanism of Peptide A cytotoxicity is generation of reactive oxygen species, which recently have been recognized as a major mode of action of important cancer drugs. Thus, this paper provides strong evidence that targeting an important AR-regulated gene is a new paradigm for effective prostate cancer therapy.
Collapse
Affiliation(s)
- Shuai Gao
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Chen-Lin Hsieh
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Meenakshi Bhansali
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Archana Kannan
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Lirim Shemshedini
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
The vast majority of all agents used to directly kill cancer cells (ionizing radiation, most chemotherapeutic agents and some targeted therapies) work through either directly or indirectly generating reactive oxygen species that block key steps in the cell cycle. As mesenchymal cancers evolve from their epithelial cell progenitors, they almost inevitably possess much-heightened amounts of antioxidants that effectively block otherwise highly effective oxidant therapies. Also key to better understanding is why and how the anti-diabetic drug metformin (the world's most prescribed pharmaceutical product) preferentially kills oxidant-deficient mesenchymal p53− −cells. A much faster timetable should be adopted towards developing more new drugs effective against p53− − cancers.
Collapse
Affiliation(s)
- Jim Watson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA.
| |
Collapse
|
42
|
Oxidative stress and cancer: an overview. Ageing Res Rev 2013; 12:376-90. [PMID: 23123177 DOI: 10.1016/j.arr.2012.10.004] [Citation(s) in RCA: 920] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/16/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022]
Abstract
Reactive species, which mainly include reactive oxygen species (ROS), are products generated as a consequence of metabolic reactions in the mitochondria of eukaryotic cells. In normal cells, low-level concentrations of these compounds are required for signal transduction before their elimination. However, cancer cells, which exhibit an accelerated metabolism, demand high ROS concentrations to maintain their high proliferation rate. Different ways of developing ROS resistance include the execution of alternative pathways, which can avoid large amounts of ROS accumulation without compromising the energy demand required by cancer cells. Examples of these processes include the guidance of the glycolytic pathway into the pentose phosphate pathway (PPP) and/or the generation of lactate instead of employing aerobic respiration in the mitochondria. Importantly, ROS levels can be used as a thermostat to monitor the damage that cells can bear. The implications for ROS regulation are highly significant for cancer therapy because commonly used radio- and chemotherapeutic drugs influence tumor outcome through ROS modulation. Moreover, the discovery of novel biomarkers that are able to predict the clinical response to pro-oxidant therapies is a crucial challenge to overcome to allow for the personalization of cancer therapies.
Collapse
|
43
|
Guan X, Liu Z, Wang L, Wang LE, Sturgis EM, Wei Q. Functional repeats (TGYCC)n in the p53-inducible gene 3 (PIG3) promoter and susceptibility to squamous cell carcinoma of the head and neck. Carcinogenesis 2012; 34:812-7. [PMID: 23241165 DOI: 10.1093/carcin/bgs388] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A polymorphic pentanucleotide microsatellite sequence (TGYCC)n within the p53-inducible gene 3 (PIG3) promoter is correlated with the extent of transcriptional activation by p53 and thought to modulate susceptibility to cancer. Using a PCR-silver staining-based single-strand conformation assay, we visualized variant genotypes of the PIG3 promoter (TGYCC)n motif in a subset of 100 subjects for each of four ethnic groups: non-Hispanic whites, African Americans, Hispanic Americans and Native Chinese. We found that PIG3 (TGYCC)15 was the most common allele but less frequent in non-Hispanic whites (0.660) than in Chinese (0.785) (P = 0.016). In an additional study of 616 patients with squamous cell carcinoma of the head and neck (SCCHN) and 623 cancer-free controls in a non-Hispanic white population, we found that compared with those who were PIG3 (TGYCC)15 homozygotes, subjects without the PIG3 (TGYCC)15 allele had a significantly increased SCCHN risk [adjusted odds ratio (OR) = 1.34; 95% CI = 1.04-1.73 for heterozygotes and OR = 1.69; 95% CI = 1.18-2.44 for variant homozygotes] in an allele-dose response manner (P = 0.002). Consistently, subsequent luciferase reporter assay revealed that the wild-type (TGYCC)15 allele had the highest p53-mediated transcriptional activity, compared with the other (TGYCC)n motifs. Our data suggest that the PIG3 variant polymorphic repeats alleles other than (TGYCC)15 may affect p53 binding and thus may be a marker for susceptibility to SCCHN, but our findings need to be validated in larger studies.
Collapse
Affiliation(s)
- Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | | | | | | | | | | |
Collapse
|
44
|
Zhu YY, Yu G, Zhang Y, Xu Z, Wang YQ, Yan GR, He QY. A novel andrographolide derivative AL-1 exerts its cytotoxicity on K562 cells through a ROS-dependent mechanism. Proteomics 2012; 13:169-78. [DOI: 10.1002/pmic.201200273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 10/23/2012] [Accepted: 10/25/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Yong-Yang Zhu
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| | - Guangchuang Yu
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| | - Ye Zhang
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| | - Zheng Xu
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| | - Yu-Qiang Wang
- Institute of New Drug Research; College of Pharmacy; Jinan University; Guangzhou China
| | - Guang-Rong Yan
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| | - Qing-Yu He
- Institute of Life and Health Engineering; and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes; Jinan University; Guangzhou China
| |
Collapse
|
45
|
Monument MJ, Johnson KM, Grossmann AH, Schiffman JD, Randall RL, Lessnick SL. Microsatellites with macro-influence in ewing sarcoma. Genes (Basel) 2012; 3:444-60. [PMID: 24704979 PMCID: PMC3899989 DOI: 10.3390/genes3030444] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/04/2012] [Accepted: 07/05/2012] [Indexed: 01/02/2023] Open
Abstract
Numerous molecular abnormalities contribute to the genetic derangements involved in tumorigenesis. Chromosomal translocations are a frequent source of these derangements, producing unique fusion proteins with novel oncogenic properties. EWS/ETS fusions in Ewing sarcoma are a prime example of this, resulting in potent chimeric oncoproteins with novel biological properties and a unique transcriptional signature essential for oncogenesis. Recent evidence demonstrates that EWS/FLI, the most common EWS/ETS fusion in Ewing sarcoma, upregulates gene expression using a GGAA microsatellite response element dispersed throughout the human genome. These GGAA microsatellites function as enhancer elements, are sites of epigenetic regulation and are necessary for EWS/FLI DNA binding and upregulation of principal oncogenic targets. An increasing number of GGAA motifs appear to substantially enhance EWS/FLI-mediated gene expression, which has compelling biological implications as these GGAA microsatellites are highly polymorphic within and between ethnically distinct populations. Historically regarded as junk DNA, this emerging evidence clearly demonstrates that microsatellite DNA plays an instrumental role in EWS/FLI-mediated transcriptional regulation and oncogenesis in Ewing sarcoma. This unprecedented role of GGAA microsatellite DNA in Ewing sarcoma provides a unique opportunity to expand our mechanistic understanding of how EWS/ETS fusions influence cancer susceptibility, prognosis and transcriptional regulation.
Collapse
Affiliation(s)
- Michael J Monument
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| | - Kirsten M Johnson
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| | - Allie H Grossmann
- Department of Pathology and Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA.
| | - Joshua D Schiffman
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| | - R Lor Randall
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| | - Stephen L Lessnick
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
46
|
Oxidatively generated complex DNA damage: tandem and clustered lesions. Cancer Lett 2012; 327:5-15. [PMID: 22542631 DOI: 10.1016/j.canlet.2012.04.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/02/2012] [Accepted: 04/15/2012] [Indexed: 11/22/2022]
Abstract
There is an increasing interest for oxidatively generated complex lesions that are potentially more detrimental than single oxidized nucleobases. In this survey, the recently available information on the formation and processing of several classes of complex DNA damage formed upon one radical hit including mostly hydroxyl radical and one-electron oxidants is critically reviewed. The modifications include tandem base lesions, DNA-protein cross-links and intrastrand (purine 5',8-cyclonucleosides, adjacent base cross-links) and interstrand cross-links. Information is also provided on clustered lesions produced essentially by exposure of cells to ionizing radiation and high energetic heavy ions through the involvement of multiple radical events that induce several lesions DNA in a close spatial vicinity. These consist mainly of double strand breaks (DSBs) and non-DSB clustered lesions that are referred as to oxidatively generated clustered DNA lesions (OCDLs).
Collapse
|