1
|
Das S, Dey A, Maji S, Sahoo A, Barman A, Santra S, Mondal S, Mondal KC, Chattopadhyay S. Attenuation of fluoride-induced hepatorenal oxidative stress by ferulic acid in vivo: An approach with in-silico analysis and interaction informatics of ferulic acid. J Trace Elem Med Biol 2023; 77:127133. [PMID: 36638706 DOI: 10.1016/j.jtemb.2023.127133] [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: 03/31/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Chronic fluoride toxicity induces oxidative strain and lipid peroxidation and imparts deleterious effects on human metabolic organs. AIM The present study aimed to expose the defensive impact of ferulic acid against sodium fluoride (NaF) induced hepatorenal dysfunction at the biochemical and antioxidative systems. METHODS In-vivo. Rats were arbitrarily separated into five groups as control, sodium fluoride-treated (200 ppm kg -1), vitamin C -as a positive control, and FA co-administered groups with 10 mg kg -1 and 20 mg kg -1 body weight for 56 days. In the present investigation, we measured antioxidant enzymes, superoxide dismutase, catalase, and lactate dehydrogenase by electrozymographic and spectrophotometric methods. Biochemical assessment of TBARS, conjugated diene, and different serum biomarkers was done for liver and kidney functionality tests. In-silico. An in-silico study was conducted through a molecular docking experiment to evaluate the binding potentiality of FA by employing AutoDock Vina [version 1.5.6] to overcome the abnormality in the activities of catalase, and superoxide dismutase in NaF promoted toxicity of hepatorenal system. In-vitro. An in vitro biochemical experiment was conducted to support the in-silico study. RESULTS Superoxide dismutase and catalase were decreased in the intoxicated rat. Ferulic acid (FA) as an antioxidant remarkably defended the NaF-mediated deterioration of the antioxidative status in the hepatorenal system, lowering lipid peroxidation products, malondialdehyde, and conjugated diene. Serum biomarkers, ALT, AST, ALP, urea, and creatinine increased in the intoxicated group than in control. Ferulic acid significantly neutralized the ill effects of NaF on serum lipid profile. In-silico analysis hypothesized the strong interaction of FA with the active side of catalase and superoxide dismutase that prevented the binding of NaF at the active site of these mentioned enzymes and this was further validated by in-vitro assay. CONCLUSION However, FA modulates free radical generation and protected these metabolic organs against sodium fluoride-induced injury.
Collapse
Affiliation(s)
- Seba Das
- Centre for Life Sciences, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Arindam Dey
- Dept. of Biomedical Laboratory Science and Management and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Shilpa Maji
- Dept. of Biomedical Laboratory Science and Management and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Asim Sahoo
- Centre for Life Sciences, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Ananya Barman
- Dept. of Biomedical Laboratory Science and Management and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Sourav Santra
- Dept. of Microbiology, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Subhadeep Mondal
- Centre for Life Sciences, Vidyasagar University, Midnapore 721102, West Bengal, India
| | | | - Sandip Chattopadhyay
- Centre for Life Sciences, Vidyasagar University, Midnapore 721102, West Bengal, India; Dept. of Biomedical Laboratory Science and Management and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore 721102, West Bengal, India.
| |
Collapse
|
2
|
Didier AJ, Stiene J, Fang L, Watkins D, Dworkin LD, Creeden JF. Antioxidant and Anti-Tumor Effects of Dietary Vitamins A, C, and E. Antioxidants (Basel) 2023; 12:632. [PMID: 36978880 PMCID: PMC10045152 DOI: 10.3390/antiox12030632] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Oxidative stress, a condition characterized by an imbalance between pro-oxidant molecules and antioxidant defense systems, is increasingly recognized as a key contributor to cancer development. This is because the reactive oxygen species (ROS) generated during oxidative stress can damage DNA, proteins, and lipids to facilitate mutations and other cellular changes that promote cancer growth. Antioxidant supplementation is a potential strategy for decreasing cancer incidence; by reducing oxidative stress, DNA damage and other deleterious cellular changes may be attenuated. Several clinical trials have been conducted to investigate the role of antioxidant supplements in cancer prevention. Some studies have found that antioxidant supplements, such as vitamin A, vitamin C, and vitamin E, can reduce the risk of certain types of cancer. On the other hand, some studies posit an increased risk of cancer with antioxidant supplement use. In this review, we will provide an overview of the current understanding of the role of oxidative stress in cancer formation, as well as the potential benefits of antioxidant supplementation in cancer prevention. Additionally, we will discuss both preclinical and clinical studies highlighting the potentials and limitations of preventive antioxidant strategies.
Collapse
Affiliation(s)
- Alexander J. Didier
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | | | | | | | | | - Justin F. Creeden
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
3
|
Puente-Cobacho B, Varela-López A, Quiles JL, Vera-Ramirez L. Involvement of redox signalling in tumour cell dormancy and metastasis. Cancer Metastasis Rev 2023; 42:49-85. [PMID: 36701089 PMCID: PMC10014738 DOI: 10.1007/s10555-022-10077-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023]
Abstract
Decades of research on oncogene-driven carcinogenesis and gene-expression regulatory networks only started to unveil the complexity of tumour cellular and molecular biology. This knowledge has been successfully implemented in the clinical practice to treat primary tumours. In contrast, much less progress has been made in the development of new therapies against metastasis, which are the main cause of cancer-related deaths. More recently, the role of epigenetic and microenviromental factors has been shown to play a key role in tumour progression. Free radicals are known to communicate the intracellular and extracellular compartments, acting as second messengers and exerting a decisive modulatory effect on tumour cell signalling. Depending on the cellular and molecular context, as well as the intracellular concentration of free radicals and the activation status of the antioxidant system of the cell, the signalling equilibrium can be tilted either towards tumour cell survival and progression or cell death. In this regard, recent advances in tumour cell biology and metastasis indicate that redox signalling is at the base of many cell-intrinsic and microenvironmental mechanisms that control disseminated tumour cell fate and metastasis. In this manuscript, we will review the current knowledge about redox signalling along the different phases of the metastatic cascade, including tumour cell dormancy, making emphasis on metabolism and the establishment of supportive microenvironmental connections, from a redox perspective.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain. .,Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
4
|
Al Khamici H, Sanchez VC, Yan H, Cataisson C, Michalowski AM, Yang HH, Li L, Lee MP, Huang J, Yuspa SH. The oxidoreductase CLIC4 is required to maintain mitochondrial function and resistance to exogenous oxidants in breast cancer cells. J Biol Chem 2022; 298:102275. [PMID: 35863434 PMCID: PMC9418444 DOI: 10.1016/j.jbc.2022.102275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
The chloride intracellular channel-4 (CLIC4) is one of the six highly conserved proteins in the CLIC family that share high structural homology with GST-omega in the GST superfamily. While CLIC4 is a multifunctional protein that resides in multiple cellular compartments, the discovery of its enzymatic glutaredoxin-like activity in vitro suggested that it could function as an antioxidant. Here, we found that deleting CLIC4 from murine 6DT1 breast tumor cells using CRISPR enhanced the accumulation of reactive oxygen species (ROS) and sensitized cells to apoptosis in response to H2O2 as a ROS-inducing agent. In intact cells, H2O2 increased the expression of both CLIC4 mRNA and protein. In addition, increased superoxide production in 6DT1 cells lacking CLIC4 was associated with mitochondrial hyperactivity including increased mitochondrial membrane potential and mitochondrial organelle enlargement. In the absence of CLIC4, however, H2O2-induced apoptosis was associated with low expression and degradation of the antiapoptotic mitochondrial protein Bcl2 and the negative regulator of mitochondrial ROS, UCP2. Furthermore, transcriptomic profiling of H2O2-treated control and CLIC4-null cells revealed upregulation of genes associated with ROS-induced apoptosis and downregulation of genes that sustain mitochondrial functions. Accordingly, tumors that formed from transplantation of CLIC4-deficient 6DT1 cells were highly necrotic. These results highlight a critical role for CLIC4 in maintaining redox-homeostasis and mitochondrial functions in 6DT1 cells. Our findings also raise the possibility of targeting CLIC4 to increase cancer cell sensitivity to chemotherapeutic drugs that are based on elevating ROS in cancer cells.
Collapse
Affiliation(s)
- Heba Al Khamici
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Vanesa C Sanchez
- Office of Science, Division of Nonclinical Science, Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hualong Yan
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Luowei Li
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Jing Huang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA.
| |
Collapse
|
5
|
Jiang H, Kimura T, Hai H, Yamamura R, Sonoshita M. Drosophila as a toolkit to tackle cancer and its metabolism. Front Oncol 2022; 12:982751. [PMID: 36091180 PMCID: PMC9458318 DOI: 10.3389/fonc.2022.982751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is one of the most severe health problems worldwide accounting for the second leading cause of death. Studies have indicated that cancers utilize different metabolic systems as compared with normal cells to produce extra energy and substances required for their survival, which contributes to tumor formation and progression. Recently, the fruit fly Drosophila has been attracting significant attention as a whole-body model for elucidating the cancer mechanisms including metabolism. This tiny organism offers a valuable toolkit with various advantages such as high genetic conservation and similar drug response to mammals. In this review, we introduce flies modeling for cancer patient genotypes which have pinpointed novel therapeutic targets and drug candidates in the salivary gland, thyroid, colon, lung, and brain. Furthermore, we introduce fly models for metabolic diseases such as diabetes mellitus, obesity, and cachexia. Diabetes mellitus and obesity are widely acknowledged risk factors for cancer, while cachexia is a cancer-related metabolic condition. In addition, we specifically focus on two cancer metabolic alterations: the Warburg effect and redox metabolism. Indeed, flies proved useful to reveal the relationship between these metabolic changes and cancer. Such accumulating achievements indicate that Drosophila offers an efficient platform to clarify the mechanisms of cancer as a systemic disease.
Collapse
Affiliation(s)
- Hui Jiang
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Taku Kimura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Oral Diagnosis and Medicine, Graduate school of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Han Hai
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- *Correspondence: Ryodai Yamamura, ; Masahiro Sonoshita,
| | - Masahiro Sonoshita
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- *Correspondence: Ryodai Yamamura, ; Masahiro Sonoshita,
| |
Collapse
|
6
|
SOD2, a Potential Transcriptional Target Underpinning CD44-Promoted Breast Cancer Progression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030811. [PMID: 35164076 PMCID: PMC8839817 DOI: 10.3390/molecules27030811] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
Abstract
CD44, a cell-adhesion molecule has a dual role in tumor growth and progression; it acts as a tumor suppressor as well as a tumor promoter. In our previous work, we developed a tetracycline-off regulated expression of CD44's gene in the breast cancer (BC) cell line MCF-7 (B5 clone). Using cDNA oligo gene expression microarray, we identified SOD2 (superoxide dismutase 2) as a potential CD44-downstream transcriptional target involved in BC metastasis. SOD2 gene belongs to the family of iron/manganese superoxide dismutase family and encodes a mitochondrial protein. SOD2 plays a role in cell proliferation and cell invasion via activation of different signaling pathways regulating angiogenic abilities of breast tumor cells. This review will focus on the findings supporting the underlying mechanisms associated with the oncogenic potential of SOD2 in the onset and progression of cancer, especially in BC and the potential clinical relevance of its various inhibitors.
Collapse
|
7
|
Park C, Lee WS, Go SI, Jeong SH, Yoo J, Cha HJ, Lee YJ, Kim HS, Leem SH, Kim HJ, Kim GS, Hong SC, Choi YH. Apoptotic Effects of Anthocyanins from Vitis coignetiae Pulliat Are Enhanced by Augmented Enhancer of the Rudimentary Homolog (ERH) in Human Gastric Carcinoma MKN28 Cells. Int J Mol Sci 2021; 22:3030. [PMID: 33809701 PMCID: PMC8002340 DOI: 10.3390/ijms22063030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 11/17/2022] Open
Abstract
Evidence suggests that augmented expression of a certain gene can influence the efficacy of targeted and conventional chemotherapies. Here, we tested whether the high expression of enhancer of the rudimentary homolog (ERH), which serves as a prognostic factor in some cancers, can influence the efficacy of anthocyanins isolated from fruits of Vitis coignetiae Pulliat, Meoru in Korea (AIMs) on human gastric cancer cells. The anticancer efficacy of AIMs was augmented in ERH-transfected MKN28 cells (E-MKN28 cells). Molecularly, ERH augmented AIM-induced caspase-dependent apoptosis by activating caspase-3 and -9. The ERH-augmented apoptotic effect was related to mitochondrial depolarization and inhibition of antiapoptotic proteins, XIAP, and Bcl-2. In addition, reactive oxygen species (ROS) generation was augmented in AIMs-treated E-MKN28 cells compared to AIMs-treated naïve MKN28 cells. In conclusion, ERH augmented AIM-induced caspase-dependent mitochondrial-related apoptosis in MKN28 cells. A decrease in expression of Bcl-2 and subsequent excessive ROS generation would be the mechanism for ERH-augmented mitochondrial-related apoptosis in AIMs-treated MKN28 cells. A decrease in expression of XIAP would be another mechanism for ERH-augmented caspase-dependent apoptosis in AIMs-treated MKN28 cells.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, Busan 47340, Korea;
| | - Won Sup Lee
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea;
| | - Se-Il Go
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea;
| | - Sang-Ho Jeong
- Departments of Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (S.-H.J.); (Y.-J.L.); (S.-C.H.)
| | - Jiyun Yoo
- Department of Microbiology/Research Institute of Life Science, College of Natural Sciences, Jinju 660-701, Korea;
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea;
| | - Young-Joon Lee
- Departments of Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (S.-H.J.); (Y.-J.L.); (S.-C.H.)
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea;
| | - Sun-Hee Leem
- Departments of Biology and Biomedical Science, Dong-A University, Busan 49315, Korea;
| | - Hye Jung Kim
- Departments of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea;
| | - Gon Sup Kim
- School of Veterinary Medicine, Division of Applied Life Science (BK 21 Program), Gyeongsang National University, Jinju 660-701, Korea;
| | - Soon-Chan Hong
- Departments of Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (S.-H.J.); (Y.-J.L.); (S.-C.H.)
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| |
Collapse
|
8
|
Evaluation of the In Vitro Cytotoxic Activity of Caffeic Acid Derivatives and Liposomal Formulation against Pancreatic Cancer Cell Lines. MATERIALS 2020; 13:ma13245813. [PMID: 33352809 PMCID: PMC7766656 DOI: 10.3390/ma13245813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer belongs to the most aggressive group of cancers, with very poor prognosis. Therefore, there is an important need to find more potent drugs that could deliver an improved therapeutic approach. In the current study we searched for selective and effective caffeic acid derivatives. For this purpose, we analyzed twelve compounds and evaluated their in vitro cytotoxic activity against two human pancreatic cancer cell lines, along with a control, normal fibroblast cell line, by the classic MTT assay. Six out of twelve tested caffeic acid derivatives showed a desirable effect. To improve the therapeutic efficacy of such active compounds, we developed a formulation where caffeic acid derivative (7) was encapsulated into liposomes composed of soybean phosphatidylcholine and DSPE-PEG2000. Subsequently, we analyzed the properties of this formulation in terms of basic physical parameters (such as size, zeta potential, stability at 4 °C and morphology), hemolytic and cytotoxic activity and cellular uptake. Overall, the liposomal formulation was found to be stable, non-hemolytic and had activity against pancreatic cancer cells (IC50 19.44 µM and 24.3 µM, towards AsPC1 and BxPC3 cells, respectively) with less toxicity against normal fibroblasts. This could represent a promising alternative to currently available treatment options.
Collapse
|
9
|
Role of Nrf2 and mitochondria in cancer stem cells; in carcinogenesis, tumor progression, and chemoresistance. Biochimie 2020; 179:32-45. [PMID: 32946993 DOI: 10.1016/j.biochi.2020.09.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/05/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are rare sub-population in tumor mass with self-renewal and differentiation abilities; CSCs are considered as the main cells which are responsible for tumor metastasis, cancer recurrence, and chemo/radio-resistance. CSCs are believed to contain low mitochondria in quantity, high concentration of nuclear factor erythroid 2-related factor 2 (Nrf2), and low reactive oxygen species (ROS) levels. Mitochondria regulate certain cellular functions, including controlling of cellular energetics, calcium signaling, cell growth and cell differentiation, cell cycle regulation, and cell death. Also, mitochondria are the main sources of intrinsic ROS production. Dysfunction of CSCs mitochondria due to oxidative phosphorylation is reported in several pathological conditions, including metabolic disorders, age-related diseases, and various types of cancers. ROS levels play a significant role in cellular signal transduction and CSCs' identity and differentiation capability. Nrf2 is a master transcription factor that plays critical functions in maintaining cellular redox hemostasis by regulating several antioxidant and detoxification pathways. Recently, the critical function of Nrf2 in CSCs has been revealed by several studies. Nrf2 is an essential molecule in the maintenance of CSCs' stemness and self-renewal in response to different oxidative stresses such as chemotherapy-induced elevation of ROS. Nrf2 enables these cells to recover from chemotherapy damages, and promotes establishment of invasion and dissemination. In this study, we have summarized the role of Nrf2 and mitochondria function CSCs, which promote cancer development. The significant role of Nrf2 in the regulation of mitochondrial function and ROS levels suggests this molecule as a potential target to eradicate CSCs.
Collapse
|
10
|
Hu X, Dong D, Xia M, Yang Y, Wang J, Su J, Sun L, Yu H. Oxidative stress and antioxidant capacity: development and prospects. NEW J CHEM 2020. [DOI: 10.1039/d0nj02041a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Signaling pathways regulating redox reactions are activated to balance the redox status and maintain the normal function of cells.
Collapse
Affiliation(s)
- Xiaoqing Hu
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Delu Dong
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Meihui Xia
- The First Hospital of Jilin University
- Changchun 130021
- P. R. China
| | - Yimeng Yang
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Jiabin Wang
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Jing Su
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Liankun Sun
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| | - Huimei Yu
- Key Laboratory of Pathobiology
- Ministry of Education
- Department of Pathophysiology
- College of Basic Medical Sciences
- Jilin University
| |
Collapse
|
11
|
Sabry SA, El-Senduny FF, Abousamra NK, Salah El-Din M, Youssef MM. Oxidative stress in CLL patients leads to activation of Th9 cells: an experimental and comprehensive survey. Immunol Med 2019; 43:36-46. [PMID: 31829825 DOI: 10.1080/25785826.2019.1700747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Older adults are mostly affected by chronic lymphocytic leukemia (CLL). The present study aimed to evaluate oxidative stress in CLL and to assess its impact on IL-9, Th9 cells levels and prognosis of cases. Seventy Egyptian CLL patients and 15 healthy controls were included. Th9 cell and immunophenotyping of abnormal B cells were assessed by flow cytometry, IL-9 level using ELISA, IL-9 mRNA by qRT-PCR, cytogenetics using FISH, and oxidative stress parameters were determined spectrophotometrically and with native gel electrophoresis. Oxidative stress was elevated in CLL that correlated with abnormal immunophenotyping, cytogenetic changes, bad prognosis, Th9 cells, and overexpression of IL-9. Levels of IL-9 and Th9 cells were strongly correlated with oxidative stress and bad prognostic markers in CLL, indicating that these cells may contribute to CLL by novel mechanisms that could include oxidant injury.
Collapse
Affiliation(s)
- Sabry A Sabry
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Fardous F El-Senduny
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Nashwa K Abousamra
- Clinical Pathology Department, Hematology Unit, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Manal Salah El-Din
- Medical Oncology Department, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Magdy M Youssef
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
12
|
R. Babu K, Tay Y. The Yin-Yang Regulation of Reactive Oxygen Species and MicroRNAs in Cancer. Int J Mol Sci 2019; 20:ijms20215335. [PMID: 31717786 PMCID: PMC6862169 DOI: 10.3390/ijms20215335] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 01/17/2023] Open
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing chemical species formed as a by-product of normal aerobic respiration and also from a number of other cellular enzymatic reactions. ROS function as key mediators of cellular signaling pathways involved in proliferation, survival, apoptosis, and immune response. However, elevated and sustained ROS production promotes tumor initiation by inducing DNA damage or mutation and activates oncogenic signaling pathways to promote cancer progression. Recent studies have shown that ROS can facilitate carcinogenesis by controlling microRNA (miRNA) expression through regulating miRNA biogenesis, transcription, and epigenetic modifications. Likewise, miRNAs have been shown to control cellular ROS homeostasis by regulating the expression of proteins involved in ROS production and elimination. In this review, we summarized the significance of ROS in cancer initiation, progression, and the regulatory crosstalk between ROS and miRNAs in cancer.
Collapse
Affiliation(s)
- Kamesh R. Babu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Yvonne Tay
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Correspondence: ; Tel.: +65-6516-7756
| |
Collapse
|
13
|
Perveen H, Dey A, Nilavar NM, Chandra GK, Islam SS, Chattopadhyay S. Dietary CCPS from bitter gourd attenuates sodium arsenite induced female reproductive ailments cum infertility in wistar rats: anti-inflammatory and anti-apoptotic role. Food Chem Toxicol 2019; 131:110545. [PMID: 31163222 DOI: 10.1016/j.fct.2019.05.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/26/2019] [Accepted: 05/29/2019] [Indexed: 11/26/2022]
Abstract
This investigation explored a dietary therapy of pectic polysaccharide (CCPS) (2 mg/ Kg BW) against female repro-toxicity and infertility triggered by sodium arsenite (As3+) (10 mg/ Kg BW) in Wistar rats. The isolated CCPS consists of D-galactose and D-methyl galacturonate with a molar ratio of 1: 4. FTIR spectral analysis of CCPS and CCPS- sodium arsenite (As3+) complex indicated a possible chelating property of CCPS in presence of binding sites (OH-/COOH) for As3+. Series of negatively charged galacturonate residues in CCPS provide better potential for cation chelation. CCPS significantly mitigated As3+ induced ovarian, uterine lipid peroxidation, and reactive oxygen species (ROS) generation by the restoration of superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx) activities. CCPS post-treatment enhanced ovarian steroidogenesis along with a restoration of normal tissue histoarchitecture in As3+ fed rats by regulating the estradiol receptor alpha (ER-α). CCPS suppressed anti-inflammatory properties effectively found since a down-regulation of NF-kappa B (NF-қB), pro-inflammatory tumor necrosis-α (TNF-α) and interleukin-6 (IL-6) were observed in arsenicated rats with CCPS. This study confirmed the up-regulation of uterine pro-apoptotic/ apoptotic proteins caspase-3, poly ADP ribose polymerase (PARP), proliferating cell nuclear antigen (PCNA), phospho p53 and Bax, followed by down-regulation of Bcl-2 and protein Kinase B (AKT) signaling pathway along with uterine tissue regeneration in As3+ exposed rats. Oral CCPS attenuated the above apoptotic expressional changes significantly and dietary CCPS ensured successful fertility with the birth of healthy pups in lieu of infertile condition in As3+ fed rats. Moreover, this study also supports that CCPS treatment attenuated the As3+ toxicity by modulating the S-adenosine methionine (SAM) pool components, B12, folate and homocysteine.
Collapse
Affiliation(s)
- Hasina Perveen
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Arindam Dey
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Namrata M Nilavar
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560 012, India
| | - Goutam Kumar Chandra
- Department of Physics, National Institute of Technology Calicut, Calicut, 673 601, Kerala, India
| | - Syed Sirajul Islam
- Department of Chemistry and Chemical Technology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
14
|
Chattopadhyay S, Khatun S, Maity M, Jana S, Perveen H, Dash M, Dey A, Jana LR, Maity PP. Association of Vitamin B 12, Lactate Dehydrogenase, and Regulation of NF-κB in the Mitigation of Sodium Arsenite-Induced ROS Generation in Uterine Tissue by Commercially Available Probiotics. Probiotics Antimicrob Proteins 2019; 11:30-42. [PMID: 28994024 DOI: 10.1007/s12602-017-9333-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Managing arsenic intoxication with conventional metal chelators is a global challenge. The present study demonstrated the therapeutic role of probiotics against arsenic-induced oxidative stress and female reproductive dysfunction. Sodium arsenite-treated (1.0 mg/100 g body weight) Wistar female rats were followed up by a post-treatment of commercially available probiotic mixture in powder form (0.25 mg/100 g body weight) orally. Rats that experienced arsenic ingestion showed a significant lessening in the activities of uterine superoxide dismutase (SOD), catalase activities, and the level of non-protein soluble thiol (NPSH) with a concomitant increase in malondialdehyde (MDA) and conjugated dienes (CD). Exposure to arsenic significantly lowered the levels of vitamin B12 and estradiol. Exposure to arsenic highly expressed the inflammatory marker and transcription factor NF-κB. Arsenic-mediated instability of these above parameters was controlled by the probiotics with a rebuilding of better function of anti-oxidant components. Besides its function in regulating endogenous anti-oxidant system, probiotics were able to augment the protection against mutagenic uterine DNA-breakage, necrosis, and ovarian-uterine tissue damages in arsenicated rats.
Collapse
Affiliation(s)
- Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India. .,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India.
| | - Shamima Khatun
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Moulima Maity
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Suryashis Jana
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Hasina Perveen
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Moumita Dash
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Arindam Dey
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Lipi Rani Jana
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Pikash Pratim Maity
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.,Clinical Nutrition and Dietetics division, (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| |
Collapse
|
15
|
Perveen H, Chattopadhyay S, Maity M, Dash M, Islam SS. Involvement of proinflammatory cytokines and metallothionein in the repairing of arsenic-mediated uterine tissue damage by curcumin. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2017-0179. [PMID: 31199764 DOI: 10.1515/jbcpp-2017-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
Background Curcumin is extensively used as a therapeutic intervention for treating several ailments. The antioxidant curcumin has an anti-inflammatory and chelating property with arsenic to exhibit a strong therapeutic effect on reproductive organs. This study was undertaken to describe the protective effect of noninvasive administration of curcumin against sodium-arsenite-mediated uterine hazards in female Wistar rats. Methods Twenty-four female Wistar rats were randomly divided into four groups. The treatment was continued for 8 days and given orally sodium arsenite (10 mg/kg body weight) in combination with curcumin (20 mg/kg body weight). Results Our evaluation revealed that 8 days of sodium arsenite (10 mg/kg body weight) treatment reduced the activities of the uterine enzymatic antioxidants superoxide dismutase, catalase, and peroxidase. Blood levels of vitamin B12 and folic acid decreased followed by an increased serum lactate dehydrogenase, homocysteine level, and hepatic metallothionein-1 in arsenic-treated rats. Necrosis of uterine tissue along with the disruption of ovarian steroidogenesis was marked in arsenic-treated rats with an upregulation of uterine NF-κB and IL-6 along with a raised level of serum TNF-α. Oral administration of curcumin (20 mg/kg body weight/day) in arsenic-treated rats significantly reinstated these alterations of the antioxidant system followed by an improvement of ovarian steroidogenesis and the circulating level of B12 and folate along with the downregulation of serum homocysteine, metallothionein-1, and cytokines. Conclusions The findings of this study clearly and strongly elucidated that arsenic-induced oxidative stress in uterus is linked to an alteration of inflammation-signaling biomarkers and these have been protected through the co-administration of curcumin due to its anti-inflammatory, free radical scavenging, and antioxidant activity by the possible regulation of an S-adenosine methionine pool.
Collapse
Affiliation(s)
- Hasina Perveen
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, UGC Innovative Department, Vidyasagar University, Midnapore, West Bengal, India
| | - Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, UGC Innovative Department, Vidyasagar University, Midnapore-721102, West Bengal, India
| | - Moulima Maity
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, UGC Innovative Department, Vidyasagar University, Midnapore, West Bengal, India
| | - Moumita Dash
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division, UGC Innovative Department, Vidyasagar University, Midnapore, West Bengal, India
| | - Syed Sirajul Islam
- Department of Chemistry and Chemical Technology, Vidyasagar University, Midnapore, West Bengal, India
| |
Collapse
|
16
|
Ding F, Sun K, Sun N, Jiang Q, Cao M, Wu Z. iTRAQ-based proteomics reveals SOD2 as a potential salivary biomarker in liver cancer. Int J Biol Markers 2019; 34:221-231. [PMID: 31041878 DOI: 10.1177/1724600819841619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Salivary proteomic analysis has been extensively used in a wide range of cancer, but not in hepatocellular carcinoma. The aim of this study was to identify potential salivary biomarkers for hepatocellular carcinoma clinical screening. METHODS In this study, we performed isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics analysis to detect differentially expressed proteins between saliva samples from 15 hepatocellular carcinoma patients and 15 healthy controls. Enzyme-linked immunosorbent assay (ELISA) verification was undertaken in saliva samples from 14 hepatocellular carcinoma patients and 14 healthy controls. RESULTS Overall, 133 proteins with significant differential expression level (ratio > 1.5 or < 0.67) were detected. Using bioinformatic analysis, two candidate proteins were selected and subsequently verified by ELISA. The increased expression of superoxide dismutase 2, mitochondrial (SOD2) in hepatocellular carcinoma patients was confirmed by ELISA, with an area under the curve value of 0.9082. CONCLUSIONS iTRAQ-based quantitative proteomics revealed that SOD2 might serve as a potential salivary biomarker for hepatocellular carcinoma detection. Our results indicated that a noninvasive and inexpensive salivary test might be established for hepatocellular carcinoma detection.
Collapse
Affiliation(s)
- Feng Ding
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Kehuan Sun
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Ningning Sun
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Qianqian Jiang
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Meiqun Cao
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Zhengzhi Wu
- Shenzhen Geriatric Research Institute, Shenzhen, Guangdong, China.,Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Chao W, Deng JS, Li PY, Kuo YH, Huang GJ. Inotilone from Inonotus linteus suppresses lung cancer metastasis in vitro and in vivo through ROS-mediated PI3K/AKT/MAPK signaling pathways. Sci Rep 2019; 9:2344. [PMID: 30787353 PMCID: PMC6382761 DOI: 10.1038/s41598-019-38959-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Metastasis is one of the main causes of mortality in cancer patients. Inotilone, a major component of Inonotus linteus, is a traditional Chinese medical herb. In this study, MTT results showed that inotilone had no obvious cytotoxicity. Animal model results revealed that inotilone suppressed cancer metastatic efficacy. Serum results showed that inotilone reduced the activity of matrix metalloproteinase (MMP)-2 and -9 and tumor necrosis factor alpha (TNF-α) activity as well as NO content. Additionally, inotilone affected MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-2 protein expression and improved the activity of the antioxidant enzymes in the lung tissues of LLC-bearing mice. In addition, cell experimental results showed that inotilone reduced the activity of MMP-2/-9 and inhibited the ability for cellular migration and invasion. Inotilone decreased interleukin (IL)-8 expression in A549 cells. Western blot results revealed that inotilone affected the protein expression of MMPs, nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, anti-oxidant enzymes, mitogen activated protein kinase (MAPK), focal adhesion kinase (FAK), phosphoinositide-3 kinase (PI3K)-AKT, and nuclear factor (NF)κB. Therefore, we propose that inotilone is a potential therapeutic candidate against metastatic lung cancer cells.
Collapse
Affiliation(s)
- Wei Chao
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jeng-Shyan Deng
- Department of Health and Nutrition Biotechnology, College of Medical and Health Science, Asia University, Taichung, 413, Taiwan
| | - Pei-Ying Li
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Yueh-Hsiung Kuo
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Guan-Jhong Huang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
18
|
Biological activity of Pt IV prodrugs triggered by riboflavin-mediated bioorthogonal photocatalysis. Sci Rep 2018; 8:17198. [PMID: 30464209 PMCID: PMC6249213 DOI: 10.1038/s41598-018-35655-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
We have recently demonstrated that riboflavin (Rf) functions as unconventional bioorthogonal photocatalyst for the activation of PtIV prodrugs. In this study, we show how the combination of light and Rf with two PtIV prodrugs is a feasible strategy for light-mediated pancreatic cancer cell death induction. In Capan-1 cells, which have high tolerance against photodynamic therapy, Rf-mediated activation of the cisplatin and carboplatin prodrugs cis,cis,trans-[Pt(NH3)2(Cl)2(O2CCH2CH2CO2H)2] (1) and cis,cis,trans-[Pt(NH3)2(CBDCA)(O2CCH2CH2CO2H)2] (2, where CBDCA = cyclobutane dicarboxylate) resulted in pronounced reduction of the cell viability, including under hypoxia conditions. Such photoactivation mode occurs to a considerable extent intracellularly, as demonstrated for 1 by uptake and cell viability experiments. 195Pt NMR, DNA binding studies using circular dichroism, mass spectrometry and immunofluorescence microscopy were performed using the Rf-1 catalyst-substrate pair and indicated that cell death is associated with the efficient light-induced formation of cisplatin. Accordingly, Western blot analysis revealed signs of DNA damage and activation of cell death pathways through Rf-mediated photochemical activation. Phosphorylation of H2AX as indicator for DNA damage, was detected for Rf-1 in a strictly light-dependent fashion while in case of free cisplatin also in the dark. Photochemical induction of nuclear pH2AX foci by Rf-1 was confirmed in fluorescence microscopy again proving efficient light-induced cisplatin release from the prodrug system.
Collapse
|
19
|
Khatun S, Maity M, Perveen H, Dash M, Chattopadhyay S. Spirulina platensis ameliorates arsenic-mediated uterine damage and ovarian steroidogenic disorder. Facets (Ott) 2018. [DOI: 10.1139/facets-2017-0099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Novel, non-invasive, painless oral therapeutic agents are needed to replace the painful conventional treatment of arsenic-associated health hazards with metal chelators. Our aim was to examine the effect of spirulina ( Spirulina platensis ( Geitler, 1925 )) on arsenic-mediated uterine toxicity. Female Wistar rats were divided equally into four experimental treatment groups: control group, sodium arsenite group (1.0 mg/100 g body mass), spirulina placebo group (20 mg/100 g body mass), and sodium arsenite + spirulina group. In contrast with the control group, spectrophotometric and electrozymographic evaluation revealed that rats that ingested arsenic for 8 d showed significant diminution of the activities of superoxide dismutase, catalase, and peroxidase ( p < 0.001). Mutagenic uterine DNA breakage and tissue damage were prominent following arsenic consumption by the rats. Oral delivery of spirulina resulted in a significant amelioration of arsenic-induced adverse oxidative stress and genotoxic state of rats. A significant low-signaling ( p < 0.001) of gonadotropins and estradiol was also noted in the arsenic-treated rats, which was terminated by spirulina; this arsenic-primed adverse effect was significant ( p < 0.05, p < 0.01). The spirulina treatment mechanism could be associated with augmentation of the antioxidant defense system that protects the arsenic-mediated pathological state of the uterus.
Collapse
Affiliation(s)
- Shamima Khatun
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal 721102, India
| | - Moulima Maity
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal 721102, India
| | - Hasina Perveen
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal 721102, India
| | - Moumita Dash
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal 721102, India
| | - Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics Division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal 721102, India
| |
Collapse
|
20
|
Affiliation(s)
- Matthew S. Alexander
- Departments of Surgery and Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa Hospitals and Clinics and the University of Iowa College of Medicine, Iowa City, IA, USA
| | - Joseph J. Cullen
- Departments of Surgery and Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa Hospitals and Clinics and the University of Iowa College of Medicine, Iowa City, IA, USA
| |
Collapse
|
21
|
Dey A, Chattopadhyay S, Jana S, Giri MK, Khatun S, Dash M, Perveen H, Maity M. Restoration of uterine redox-balance by methanolic extract of Camellia sinensis in arsenicated rats. ACTA ACUST UNITED AC 2018. [DOI: 10.14232/abs.2018.1.7-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Arsenic, an environmental and industrial pollutant causes female reproductive disturbances and female infertility. Several researchers found that the use of Camellia sinensis (CS) (green tea) is effective as an alternative therapeutic strategy in the management of several health ailments. This study explores the role of CS extract against arsenic-induced rat uterine tissue damage. Methanolic extract of CS (10 mg/kg BW) was tested concomitantly in arsenic-treated (10 mg/kg BW) rats for a duration of two-oestrous cycle length (8 days). CS effectively attenuated arsenic-induced antioxidantdepletion and necrosis in uterine tissue. Rats treated with sodium arsenite showed significantlyreduced activities of enzymatic antioxidants like superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) in uterine tissue as evidenced by the results of spectrophotometric and electrozymographic analysis. Co-administration of CS significantly reversed the above oxidative stress markers in uterine tissue along with the histopathological changes in ovarian and uterine tissue. Moreover, an increase in the level of transcription factor NF-κB in the uterine tissue in association with reduced serum levels of vitamin B12 and folic acid were mitigated in arsenic fed rats following CS co-administration.
Collapse
|
22
|
Jayakumar T, Liu CH, Wu GY, Lee TY, Manubolu M, Hsieh CY, Yang CH, Sheu JR. Hinokitiol Inhibits Migration of A549 Lung Cancer Cells via Suppression of MMPs and Induction of Antioxidant Enzymes and Apoptosis. Int J Mol Sci 2018; 19:ijms19040939. [PMID: 29565268 PMCID: PMC5979393 DOI: 10.3390/ijms19040939] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 02/04/2023] Open
Abstract
Hinokitiol, a natural monoterpenoid from the heartwood of Calocedrus formosana, has been reported to have anticancer effects against various cancer cell lines. However, the detailed molecular mechanisms and the inhibiting roles of hinokitiol on adenocarcinoma A549 cells remain to be fully elucidated. Thus, the current study was designed to evaluate the effect of hinokitiol on the migration of human lung adenocarcinoma A549 cells in vitro. The data demonstrates that hinokitiol does not effectively inhibit the viability of A549 cells at up to a 10 µM concentration. When treated with non-toxic doses (1–5 µM) of hinokitiol, the cell migration is markedly suppressed at 5 µM. Hinokitiol significantly reduced p53 expression, followed by attenuation of Bax in A549 cells. A dose-dependent inhibition of activated caspase-9 and -3 was observed in the presence of hinokitiol. An observed increase in protein expression of matrix metalloproteinases (MMPs) -2/-9 in A549 cells was significantly inhibited by hinokitiol. Remarkably, when A549 cells were subjected to hinokitiol (1–5 µM), there was an increase in the activities of antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) from the reduction in cells. In addition, the incubation of A549 cells with hinokitiol significantly activated the cytochrome c expression, which may be triggered by activation of caspase-9 followed by caspase-3. These observations indicate that hinokitiol inhibited the migration of lung cancer A549 cells through several mechanisms, including the activation of caspases-9 and -3, induction of p53/Bax and antioxidant CAT and SOD, and reduction of MMP-2 and -9 activities. It also induces cytochrome c expression. These findings demonstrate a new therapeutic potential for hinokitiol in lung cancer chemoprevention.
Collapse
Affiliation(s)
- Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chao-Hong Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Dermatology, Yuan's General Hospital, Kaohsiung 249, Taiwan.
| | - Guan-Yi Wu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Tzu-Yin Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Manjunath Manubolu
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH 43212, USA.
| | - Cheng-Ying Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
23
|
Maity M, Perveen H, Dash M, Jana S, Khatun S, Dey A, Mandal AK, Chattopadhyay S. Arjunolic Acid Improves the Serum Level of Vitamin B 12 and Folate in the Process of the Attenuation of Arsenic Induced Uterine Oxidative Stress. Biol Trace Elem Res 2018; 182:78-90. [PMID: 28660490 DOI: 10.1007/s12011-017-1077-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 06/08/2017] [Indexed: 01/13/2023]
Abstract
Continuation of prolonged treatment against arsenicosis with conventional chelating therapy is a global challenge. The present study was intended to evaluate the defensive effect of arjunolic acid against arsenic-induced oxidative stress and female reproductive dysfunction. Wistar strain adult female rats were given sodium arsenite (10 mg/kg body weight) in combination with arjunolic acid (10 mg/kg body weight) orally for two estrous cycles. Electrozymographic analysis explored that arjunolic acid co-treatment counteracted As3+-induced ROS production in uterine tissue by stimulating the activities of endogenous enzymatic antioxidants. Arjunolic acid was able to enhance the protection against mutagenic uterine DNA breakage, necrosis, and ovarian-uterine tissue damages in arsenicated rats by improving the ovarian steroidogenesis. The mechanisms might be coupled with the augmentation of antioxidant defense system, partly through the elimination of arsenic with the involvement of S-adenosyl methionine pool where circulating levels of vitamin B12, folic acid, and homocysteine play critical roles as evidenced from our present investigation.
Collapse
Affiliation(s)
- Moulima Maity
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Hasina Perveen
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Moumita Dash
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Suryashis Jana
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Shamima Khatun
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Arindam Dey
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Amit Kumar Mandal
- Department of Microbiology, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University, Uttar Dinajpur, West Bengal, 733134, India
| | - Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, Vidyasagar University, Midnapore, West Bengal, 721102, India.
- Clinical Nutrition and Dietetics Division (Funded under UGC Innovative Programme), Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
24
|
Kumari S, Badana AK, G MM, G S, Malla R. Reactive Oxygen Species: A Key Constituent in Cancer Survival. Biomark Insights 2018; 13:1177271918755391. [PMID: 29449774 PMCID: PMC5808965 DOI: 10.1177/1177271918755391] [Citation(s) in RCA: 466] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 12/30/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cancer is one of the major heterogeneous disease with high morbidity and mortality with poor prognosis. Elevated levels of reactive oxygen species (ROS), alteration in redox balance, and deregulated redox signaling are common hallmarks of cancer progression and resistance to treatment. Mitochondria contribute mainly in the generation of ROS during oxidative phosphorylation. Elevated levels of ROS have been detected in cancers cells due to high metabolic activity, cellular signaling, peroxisomal activity, mitochondrial dysfunction, activation of oncogene, and increased enzymatic activity of oxidases, cyclooxygenases, lipoxygenases, and thymidine phosphorylases. Cells maintain intracellular homeostasis by developing an immense antioxidant system including catalase, superoxide dismutase, and glutathione peroxidase. Besides these enzymes exist an important antioxidant glutathione and transcription factor Nrf2 which contribute in balancing oxidative stress. Reactive oxygen species-mediated signaling pathways activate pro-oncogenic signaling which eases in cancer progression, angiogenesis, and survival. Concomitantly, to maintain ROS homeostasis and evade cancer cell death, an increased level of antioxidant capacity is associated with cancer cells. CONCLUSIONS This review focuses the role of ROS in cancer survival pathways and importance of targeting the ROS signal involved in cancer development, which is a new strategy in cancer treatment.
Collapse
Affiliation(s)
- Seema Kumari
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| | - Anil Kumar Badana
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| | - Murali Mohan G
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| | - Shailender G
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| | - RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, India
| |
Collapse
|
25
|
Insights into the Dichotomous Regulation of SOD2 in Cancer. Antioxidants (Basel) 2017; 6:antiox6040086. [PMID: 29099803 PMCID: PMC5745496 DOI: 10.3390/antiox6040086] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
While loss of antioxidant expression and the resultant oxidant-dependent damage to cellular macromolecules is key to tumorigenesis, it has become evident that effective oxidant scavenging is conversely necessary for successful metastatic spread. This dichotomous role of antioxidant enzymes in cancer highlights their context-dependent regulation during different stages of tumor development. A prominent example of an antioxidant enzyme with such a dichotomous role and regulation is the mitochondria-localized manganese superoxide dismutase SOD2 (MnSOD). SOD2 has both tumor suppressive and promoting functions, which are primarily related to its role as a mitochondrial superoxide scavenger and H₂O₂ regulator. However, unlike true tumor suppressor- or onco-genes, the SOD2 gene is not frequently lost, or rarely mutated or amplified in cancer. This allows SOD2 to be either repressed or activated contingent on context-dependent stimuli, leading to its dichotomous function in cancer. Here, we describe some of the mechanisms that underlie SOD2 regulation in tumor cells. While much is known about the transcriptional regulation of the SOD2 gene, including downregulation by epigenetics and activation by stress response transcription factors, further research is required to understand the post-translational modifications that regulate SOD2 activity in cancer cells. Moreover, future work examining the spatio-temporal nature of SOD2 regulation in the context of changing tumor microenvironments is necessary to allows us to better design oxidant- or antioxidant-based therapeutic strategies that target the adaptable antioxidant repertoire of tumor cells.
Collapse
|
26
|
Wilkes JG, Alexander MS, Cullen JJ. Superoxide Dismutases in Pancreatic Cancer. Antioxidants (Basel) 2017; 6:antiox6030066. [PMID: 28825637 PMCID: PMC5618094 DOI: 10.3390/antiox6030066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/10/2017] [Accepted: 08/15/2017] [Indexed: 01/17/2023] Open
Abstract
The incidence of pancreatic cancer is increasing as the population ages but treatment advancements continue to lag far behind. The majority of pancreatic cancer patients have a K-ras oncogene mutation causing a shift in the redox state of the cell, favoring malignant proliferation. This mutation is believed to lead to nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation and superoxide overproduction, generating tumorigenic behavior. Superoxide dismutases (SODs) have been studied for their ability to manage the oxidative state of the cell by dismuting superoxide and inhibiting signals for pancreatic cancer growth. In particular, manganese superoxide dismutase has clearly shown importance in cell cycle regulation and has been found to be abnormally low in pancreatic cancer cells as well as the surrounding stromal tissue. Likewise, extracellular superoxide dismutase expression seems to favor suppression of pancreatic cancer growth. With an increased understanding of the redox behavior of pancreatic cancer and key regulators, new treatments are being developed with specific targets in mind. This review summarizes what is known about superoxide dismutases in pancreatic cancer and the most current treatment strategies to be advanced from this knowledge.
Collapse
Affiliation(s)
- Justin G. Wilkes
- Departments of Surgery and Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA 52245, USA; (J.G.W.); (M.S.A.)
| | - Matthew S. Alexander
- Departments of Surgery and Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA 52245, USA; (J.G.W.); (M.S.A.)
| | - Joseph J. Cullen
- Departments of Surgery and Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA 52245, USA; (J.G.W.); (M.S.A.)
- Veterans Affairs Medical Center, Iowa City, IA 52245, USA
- Correspondence: ; Tel.: +1-319-353-8297; Fax: +1-319-356-8378
| |
Collapse
|
27
|
Li W, Ma Z, Ma J, Li X, Xu Q, Duan W, Chen X, Lv Y, Zhou S, Wu E, Ma Q, Huo X. Hydrogen peroxide mediates hyperglycemia-induced invasive activity via ERK and p38 MAPK in human pancreatic cancer. Oncotarget 2016; 6:31119-33. [PMID: 26439801 PMCID: PMC4741592 DOI: 10.18632/oncotarget.5045] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/26/2015] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus and pancreatic cancer are intimately related, as approximately 85% of pancreatic cancer patients suffer from glucose intolerance or even diabetes. In this study, we evaluate the underlying mechanism by which hyperglycemia modulates the invasive potential of cancer cells and contributes to their enhanced metastatic behavior. Here we show that hyperglycemia increases the hydrogen peroxide (H2O2) concentration through up-regulation of manganese superoxide dismutase (SOD2) expression, which further activates the ERK and p38 MAPK pathways, as well as the transcription factors NF-κB and AP-1, in a time-dependent manner. The invasion of pancreatic cancer cells resulting from the activation of the H2O2/MAPK axis under high glucose conditions is effectively inhibited by PD 98059 (ERK inhibitor), SB 203580 (p38 MAPK inhibitor), polyethylene glycol-conjugated catalase (PEG-CAT), or the siRNA specific to SOD2. In addition, streptozotocin-treated diabetic nude mice exhibit a stronger tumor invasive ability in renal capsule xenografts which could be suppressed by PEG-CAT treatment. Furthermore, the integrated optical density (IOD) of SOD2 and uPA stainings is higher in the tumor tissues of pancreatic cancer patients with diabetes compared with pancreatic cancer patients with euglycemia. Taken together, our results demonstrate that hyperglycemia enhances cell invasive ability through the SOD2/H2O2/MAPK axis in human pancreatic cancer. Thus, SOD2/H2O2/MAPK axis may represent a promising therapeutic target for pancreatic cancer patients combined with diabetes mellitus.
Collapse
Affiliation(s)
- Wei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhenhua Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiguang Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qinhong Xu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yunfu Lv
- Department of General Surgery, People's Hospital of Hainan Province, Haikou, 570311, China
| | - Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, 58108, ND, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, 58108, ND, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiongwei Huo
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
28
|
Immunohistochemical assessment of mitochondrial superoxide dismutase (MnSOD) in colorectal premalignant and malignant lesions. GASTROENTEROLOGY REVIEW 2016; 11:239-246. [PMID: 28053678 PMCID: PMC5209461 DOI: 10.5114/pg.2016.57943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 07/20/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION It is generally accepted that mitochondria are a primary source of intracellular reactive oxygen species (ROS). Under physiological circumstances they are permanently formed as by-products of aerobic metabolism in the mitochondria. To counter the harmful effect of ROS, cells possess an antioxidant defence system to detoxify ROS and avert them from accumulation at high concentrations. Mitochondria-located manganese superoxide dismutase (MnSOD, SOD2) successfully converts superoxide to the less reactive hydrogen peroxide (H2O2). To the best of our knowledge, there are no available data regarding immunohistochemical expression of MnSOD in colorectal neoplastic tissues. AIM To investigate the immunohistochemical expression status of MnSOD in colorectal premalignant and malignant lesions. MATERIAL AND METHODS This study was performed on resected specimens obtained from 126 patients who had undergone surgical resection for primary sporadic colorectal cancer, and from 114 patients who had undergone colonoscopy at the Municipal Hospital in Jaworzno (Poland). Paraffin-embedded, 4-µm-thick tissue sections were stained for rabbit polyclonal anti SOD2 antibody obtained from GeneTex (clone TF9-10-H10 from America Diagnostica). RESULTS Results of our study demonstrated that the development of colorectal cancer is connected with increased expression of MnSOD both in adenoma and adenocarcinoma stages. Samples of adenocarcinoma with G2 and G3 grade showed significantly higher levels of immunohistochemical expression of this antioxidant enzyme. Moreover, patients with the presence of lymphovascular invasion and higher degree of regional lymph node status have been also characterised by higher levels of MnSOD expression. The samples of adenoma have been characterised by higher levels of MnSOD expression in comparison to normal mucosa as well. Interestingly, there was no significant correlation between expression and histological type of adenoma. CONCLUSIONS Development of colorectal cancer is connected with increased expression of MnSOD both in adenoma and adenocarcinoma stages.
Collapse
|
29
|
Li W, Cao L, Han L, Xu Q, Ma Q. Superoxide dismutase promotes the epithelial-mesenchymal transition of pancreatic cancer cells via activation of the H2O2/ERK/NF-κB axis. Int J Oncol 2016; 46:2613-20. [PMID: 25825208 DOI: 10.3892/ijo.2015.2938] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/03/2015] [Indexed: 01/28/2023] Open
Abstract
Our previous study revealed that superoxide dismutase (SOD)-dependent production of reactive oxygen species (ROS) was able to increase the invasive ability of pancreatic cancer cells. However, the underlying mechanisms by which SOD enhances metastasis are still not fully elucidated. As epithelial-mesenchymal transition (EMT) is a key player in tumor metastasis, the aim of this study was to evaluate whether SOD affects EMT in pancreatic cancer cells and the related mechanism. Human pancreatic cancer cells BxPC-3 and Panc-1 were utilized to examine the level of hydrogen peroxide (H2O) in the absence or presence of SOD and catalase (CAT). The activation of phospho-ERK and phospho-NF-κB were measured by western blot analysis. Wound healing assay and transwell invasion assay were used to detect the migratory and invasive potential of cancer cells. The EMT-related factors, E-cadherin, N-cadherin and vimentin were detected by QT-PCR and western blot analysis. The results of present study showed that SOD not only increased cell migration and invasion in pancreatic cancer, but also mediated the expression of EMT-related factors and cell morphology. In addition, the levels of phospho-ERK and phospho-NF-κB were induced by SOD which could be counter-balanced by both CAT treatment and PD 98059 (an ERK inhibitor). Taken together, these data indicate that SOD promotes the invasive and migratory activity of pancreatic cancer. Blocking the H2O2/ERK/NF-κB axis might be a novel strategy for the treatment of this severe malignancy.
Collapse
Affiliation(s)
- Wei Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Lei Cao
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
30
|
Pidugu LSM, Mbimba JCE, Ahmad M, Pozharski E, Sausville EA, Emadi A, Toth EA. A direct interaction between NQO1 and a chemotherapeutic dimeric naphthoquinone. BMC STRUCTURAL BIOLOGY 2016; 16:1. [PMID: 26822308 PMCID: PMC4730606 DOI: 10.1186/s12900-016-0052-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/19/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Multimeric naphthoquinones are redox-active compounds that exhibit antineoplastic, antiprotozoal, and antiviral activities. Due to their multimodal effect on perturbation of cellular oxidative state, these compounds hold great potential as therapeutic agents against highly proliferative neoplastic cells. In our previous work, we developed a series of novel dimeric naphthoquinones and showed that they were selectively cytotoxic to human acute myeloid leukemia (AML), breast and prostate cancer cell lines. We subsequently identified the oxidoreductase NAD(P)H dehydrogenase, quinone 1 (NQO1) as the major target of dimeric naphthoquinones and proposed a mechanism of action that entailed induction of a futile redox cycling. RESULTS Here, for the first time, we describe a direct physical interaction between the bromohydroxy dimeric naphthoquinone E6a and NQO1. Moreover, our studies reveal an extensive binding interface between E6a and the isoalloxazine ring of the flavin adenine dinucleotide (FAD) cofactor of NQO1 in addition to interactions with protein side chains in the active site. We also present biochemical evidence that dimeric naphthoquinones affect the redox state of the FAD cofactor of NQO1. Comparison of the mode of binding of E6a with those of other chemotherapeutics reveals unique characteristics of the interaction that can be leveraged in future drug optimization efforts. CONCLUSION The first structure of a dimeric naphthoquinone-NQO1 complex was reported, which can be used for design and synthesis of more potent next generation dimeric naphthoquinones to target NQO1 with higher affinity and specificity.
Collapse
Affiliation(s)
- Lakshmi Swarna Mukhi Pidugu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Institute for Bioscience and Biotechnology Research, and Center for Biomolecular Therapeutics, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.
| | - J C Emmanuel Mbimba
- Institute for Bioscience and Biotechnology Research, and Center for Biomolecular Therapeutics, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.
| | - Muqeet Ahmad
- Institute for Bioscience and Biotechnology Research, and Center for Biomolecular Therapeutics, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.
| | - Edwin Pozharski
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Institute for Bioscience and Biotechnology Research, and Center for Biomolecular Therapeutics, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.
| | - Edward A Sausville
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Ashkan Emadi
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Eric A Toth
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Institute for Bioscience and Biotechnology Research, and Center for Biomolecular Therapeutics, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
31
|
Jiao R, Liu Y, Gao H, Xiao J, So KF. The Anti-Oxidant and Antitumor Properties of Plant Polysaccharides. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:463-488. [DOI: 10.1142/s0192415x16500269] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Oxidative stress has been increasingly recognized as a major contributing factor in a variety of human diseases, from inflammation to cancer. Although certain parts of signaling pathways are still under investigation, detailed molecular mechanisms for the induction of diseases have been elucidated, especially the link between excessive oxygen reactive species (ROS) damage and tumorigenesis. Emerging evidence suggests anti-oxidant therapy can play a key role in treating those diseases. Among potential drug resources, plant polysaccharides are natural anti-oxidant constituents important for human health because of their long history in ethnopharmacology, wide availability and few side effects upon consumption. Plant polysaccharides have been shown to possess anti-oxidant, anti-inflammation, cell viability promotion, immune-regulation and antitumor functions in a number of disease models, both in laboratory studies and in the clinic. In this paper, we reviewed the research progress of signaling pathways involved in the initiation and progression of oxidative stress- and cancer-related diseases in humans. The natural sources, structural properties and biological actions of several common plant polysaccharides, including Lycium barbarum, Ginseng, Zizyphus Jujuba, Astragalus lentiginosus, and Ginkgo biloba are discussed in detail, with emphasis on their signaling pathways. All of the mentioned common plant polysaccharides have great potential to treat oxidative stress and cancinogenic disorders in cell models, animal disease models and clinical cases. ROS-centered pathways (e.g. mitochondrial autophagy, MAPK and JNK) and transcription factor-related pathways (e.g. NF-[Formula: see text]B and HIF) are frequently utilized by these polysaccharides with or without the further involvement of inflammatory and death receptor pathways. Some of the polysaccharides may also influence tumorigenic pathways, such as Wnt and p53 to play their anti-tumor roles. In addition, current problems and future directions for the application of those plant polysaccharides are also listed and discussed.
Collapse
Affiliation(s)
- Rui Jiao
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| | - Yingxia Liu
- State Key Discipline of Infectious Diseases, Department of Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Hao Gao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jia Xiao
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
- State Key Discipline of Infectious Diseases, Department of Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok Fai So
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- GMH Institute of Central Nervous System Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
32
|
Huang CH, Jayakumar T, Chang CC, Fong TH, Lu SH, Thomas PA, Choy CS, Sheu JR. Hinokitiol Exerts Anticancer Activity through Downregulation of MMPs 9/2 and Enhancement of Catalase and SOD Enzymes: In Vivo Augmentation of Lung Histoarchitecture. Molecules 2015; 20:17720-34. [PMID: 26404213 PMCID: PMC6332280 DOI: 10.3390/molecules201017720] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 11/29/2022] Open
Abstract
Melanoma is extremely resistant to chemotherapy and the death rate is increasing hastily worldwide. Extracellular matrix promotes the migration and invasion of tumor cells through the production of matrix metalloproteinase (MMP)-2 and -9. Evidence has shown that natural dietary antioxidants are capable of inhibiting cancer cell growth. Our recent studies showed that hinokitiol, a natural bioactive compound, inhibited vascular smooth muscle cell proliferation and platelets aggregation. The present study is to investigate the anticancer efficacy of hinokitiol against B16-F10 melanoma cells via modulating tumor invasion factors MMPs, antioxidant enzymes in vitro. An in vivo mice model of histological investigation was performed to study the patterns of elastic and collagen fibers. Hinokitiol inhibited the expression and activity of MMPs-2 and -9 in B16-F10 melanoma cells, as measured by western blotting and gelatin zymography, respectively. An observed increase in protein expression of MMPs 2/9 in melanoma cells was significantly inhibited by hinokitiol. Notably, hinokitiol (1–5 μM) increased the activities of antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) from the reduction in melanoma cells. Also, hinokitiol (2–10 µM) concentration dependently reduced in vitro Fenton reaction induced hydroxyl radical (OH·) formation. An in vivo study showed that hinokitiol treatment increased elastic fibers (EF), collagens dispersion, and improved alveolar alterations in the lungs of B16/F10 injected mice. Overall, our findings propose that hinokitiol may be a potent anticancer candidate through down regulation of MMPs 9/2, reduction of OH· production and enhancement of antioxidant enzymes SOD and CAT.
Collapse
Affiliation(s)
- Chien-Hsun Huang
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Division of Urology, Department of Surgery, Taipei City Hospital, Zhongxiao Branch, Taipei 115, Taiwan.
| | - Thanasekaran Jayakumar
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chao-Chien Chang
- Department of Cardiology, Cathay General Hospital, Taipei 106, Taiwan.
| | - Tsorng-Harn Fong
- Department of Anatomy, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei 110, Taiwan.
| | - Shing-Hwa Lu
- Division of Urology, Department of Surgery, Taipei City Hospital, Zhongxiao Branch, Taipei 115, Taiwan.
| | - Philip Aloysius Thomas
- Department of Microbiology, Institute of Ophthalmology, Joseph Eye Hospital, Tiruchirappalli 620001, Tamil Nadu, India.
| | - Cheuk-Sing Choy
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Emergency, Min-Sheng General Hospital, Taoyuan 330, Taiwan.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
33
|
Hrycay EG, Bandiera SM. Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 74:35-84. [PMID: 26233903 DOI: 10.1016/bs.apha.2015.03.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review examines the involvement of cytochrome P450 (CYP) enzymes in the formation of reactive oxygen species in biological systems and discusses the possible involvement of reactive oxygen species and CYP enzymes in cancer. Reactive oxygen species are formed in biological systems as byproducts of the reduction of molecular oxygen and include the superoxide radical anion (∙O2-), hydrogen peroxide (H2O2), hydroxyl radical (∙OH), hydroperoxyl radical (HOO∙), singlet oxygen ((1)O2), and peroxyl radical (ROO∙). Two endogenous sources of reactive oxygen species are the mammalian CYP-dependent microsomal electron transport system and the mitochondrial electron transport chain. CYP enzymes catalyze the oxygenation of an organic substrate and the simultaneous reduction of molecular oxygen. If the transfer of oxygen to a substrate is not tightly controlled, uncoupling occurs and leads to the formation of reactive oxygen species. Reactive oxygen species are capable of causing oxidative damage to cellular membranes and macromolecules that can lead to the development of human diseases such as cancer. In normal cells, intracellular levels of reactive oxygen species are maintained in balance with intracellular biochemical antioxidants to prevent cellular damage. Oxidative stress occurs when this critical balance is disrupted. Topics covered in this review include the role of reactive oxygen species in intracellular cell signaling and the relationship between CYP enzymes and cancer. Outlines of CYP expression in neoplastic tissues, CYP enzyme polymorphism and cancer risk, CYP enzymes in cancer therapy and the metabolic activation of chemical procarcinogens by CYP enzymes are also provided.
Collapse
Affiliation(s)
- Eugene G Hrycay
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada.
| | - Stelvio M Bandiera
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
O'Leary BR, Fath MA, Bellizzi AM, Hrabe JE, Button AM, Allen BG, Case AJ, Altekruse S, Wagner BA, Buettner GR, Lynch CF, Hernandez BY, Cozen W, Beardsley RA, Keene J, Henry MD, Domann FE, Spitz DR, Mezhir JJ. Loss of SOD3 (EcSOD) Expression Promotes an Aggressive Phenotype in Human Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2015; 21:1741-51. [PMID: 25634994 PMCID: PMC4383686 DOI: 10.1158/1078-0432.ccr-14-1959] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/18/2015] [Indexed: 01/05/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDA) cells are known to produce excessive amounts of reactive oxygen species (ROS), particularly superoxide, which may contribute to the aggressive and refractory nature of this disease. Extracellular superoxide dismutase (EcSOD) is an antioxidant enzyme that catalyzes the dismutation of superoxide in the extracellular environment. This study tests the hypothesis that EcSOD modulates PDA growth and invasion by modifying the redox balance in PDA. EXPERIMENTAL DESIGN We evaluated the prognostic significance of EcSOD in a human tissue microarray (TMA) of patients with PDA. EcSOD overexpression was performed in PDA cell lines and animal models of disease. The impact of EcSOD on PDA cell lines was evaluated with Matrigel invasion in combination with a superoxide-specific SOD mimic and a nitric oxide synthase (NOS) inhibitor to determine the mechanism of action of EcSOD in PDA. RESULTS Loss of EcSOD expression is a common event in PDA, which correlated with worse disease biology. Overexpression of EcSOD in PDA cell lines resulted in decreased invasiveness that appeared to be related to reactions of superoxide with nitric oxide. Pancreatic cancer xenografts overexpressing EcSOD also demonstrated slower growth and peritoneal metastasis. Overexpression of EcSOD or treatment with a superoxide-specific SOD mimic caused significant decreases in PDA cell invasive capacity. CONCLUSIONS These results support the hypothesis that loss of EcSOD leads to increased reactions of superoxide with nitric oxide, which contributes to the invasive phenotype. These results allow for the speculation that superoxide dismutase mimetics might inhibit PDA progression in human clinical disease.
Collapse
Affiliation(s)
| | - Melissa A Fath
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | | | | | - Anna M Button
- Department of Biostatistics, University of Iowa, Iowa City, Iowa
| | - Bryan G Allen
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | - Adam J Case
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | | | - Brett A Wagner
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | - Garry R Buettner
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | - Charles F Lynch
- Department of Epidemiology, University of Iowa, Iowa City, Iowa
| | | | - Wendy Cozen
- University of Southern California, Los Angeles, California
| | | | | | - Michael D Henry
- Department of Microbiology, University of Iowa, Iowa City, Iowa
| | | | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | - James J Mezhir
- Department of Surgery, University of Iowa, Iowa City, Iowa. Department of Radiation Oncology, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
35
|
Hempel N, Carrico PM, Melendez JA. Manganese superoxide dismutase (Sod2) and redox-control of signaling events that drive metastasis. Anticancer Agents Med Chem 2011; 11:191-201. [PMID: 21434856 DOI: 10.2174/187152011795255911] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 03/08/2011] [Indexed: 01/06/2023]
Abstract
Manganese superoxide dismutase (Sod2) has emerged as a key enzyme with a dual role in tumorigenic progression. Early studies were primarily directed at defining the tumor suppressive function of Sod2 based on its low level expression in many tumor types. It is now commonly held that loss of Sod2 expression is likely an early event in tumor progression allowing for further propagation of the tumorigenic phenotype resulting from steady state increases in free radical production. Increases in free radical load have also been linked to defects in mitochondrial function and metastatic disease progression. It was initially believed that Sod2 loss may propagate metastatic disease progression, in reality both epidemiologic and experimental evidence indicate that Sod2 levels increase in many tumor types as they progress from early stage non-invasive disease to late stage metastatic disease. Sod2 overexpression in many instances enhances the metastatic phenotype that is reversed by efficient H(2)O(2) scavenging. This review evaluates the many sequelae associated with increases in Sod2 that impinge on the metastatic phenotype. The ability to use Sod2 to modulate the cellular redox-environment has allowed for the identification of redox-responsive signaling events that drive malignancy, such as invasion, migration and prolonged tumor cell survival. Further studies of these redox-driven events will help in the development of targeted therapeutic strategies to efficiently restrict redox-signaling essential for malignant progression.
Collapse
Affiliation(s)
- Nadine Hempel
- Center for Immunology and Microbial Diseases, Albany Medical College, 47 New Scotland Avenue, Albany NY 12208, USA
| | | | | |
Collapse
|
36
|
Lee J, Lim KT. Inhibitory effect of phytoglycoprotein (24 kDa) on hepatocarcinogenesis in N-nitrosodiethylamine-treated ICR mice. J Pharm Pharmacol 2011; 63:840-8. [DOI: 10.1111/j.2042-7158.2011.01277.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Abstract
Objectives
Hepatocellular carcinoma (HCC) is becoming one of the most prominent types of cancer in the world. For a long time in Korea Zanthoxylum piperitum DC (ZPDC) has been used in folk medicine to cure several cancers and inflammation. This study was designed to investigate whether ZPDC glycoprotein protected liver tissues against hepatocarcinogenic compounds such as N-nitrosodiethylamine (DEN).
Methods
To study the chemopreventive effect of ZPDC glycoprotein on hepatocarcinogenesis, ICR mice were injected intraperitoneally with DEN (50 mg/kg) for four weeks. We evaluated the indicators of liver tissue damage (the activity of lactate dehydrogenase (LDH) and alanine aminotransferase (ALT), and thiobarbituric acid-reactive substances (TBARS)), antioxidative enzymes (activity of superoxide dismutase (SOD) and glutathione peroxidase (GPx)), hepatocarcinogenic indicator (heat shock protein (HSP) 70) and hepatocarcinogenic signals (activity of nuclear factor (NF)-κB, cyclooxygenase (COX)-2, and matrix metalloproteinase (MMP)-9) using biochemical methods and immunoblot analysis.
Key findings
The results obtained from this study revealed that ZPDC glycoprotein (20 mg/kg) decreased the levels of LDH, ALT, and TBARS, whereas the activity of SOD and GPx increased in the DEN-treated ICR mice. With respect to the hepatocarcinogenic indicator and hepatocarcinogenic signals, HSP70, NF-κB, COX-2, and MMP-9 activity decreased.
Conclusion
The findings suggested that ZPDC glycoprotein prevented damage to liver tissue caused by DEN in the experimental mouse model.
Collapse
Affiliation(s)
- Jin Lee
- Molecular Biochemistry Laboratory, Biotechnology Research Institute and Center for the Control of Animal Hazards Using Biotechnology (BK21), Chonnam National University, Gwang-ju, South Korea
| | - Kye-Taek Lim
- Molecular Biochemistry Laboratory, Biotechnology Research Institute, Chonnam National University, Gwang-ju, South Korea
| |
Collapse
|
37
|
Pan S, Chen R, Crispin DA, May D, Stevens T, McIntosh MW, Bronner MP, Ziogas A, Anton-Culver H, Brentnall TA. Protein alterations associated with pancreatic cancer and chronic pancreatitis found in human plasma using global quantitative proteomics profiling. J Proteome Res 2011; 10:2359-76. [PMID: 21443201 DOI: 10.1021/pr101148r] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a lethal disease that is difficult to diagnose at early stages when curable treatments are effective. Biomarkers that can improve current pancreatic cancer detection would have great value in improving patient management and survival rate. A large scale quantitative proteomics study was performed to search for the plasma protein alterations associated with pancreatic cancer. The enormous complexity of the plasma proteome and the vast dynamic range of protein concentration therein present major challenges for quantitative global profiling of plasma. To address these challenges, multidimensional fractionation at both protein and peptide levels was applied to enhance the depth of proteomics analysis. Employing stringent criteria, more than 1300 proteins total were identified in plasma across 8-orders of magnitude in protein concentration. Differential proteins associated with pancreatic cancer were identified, and their relationship with the proteome of pancreatic tissue and pancreatic juice from our previous studies was discussed. A subgroup of differentially expressed proteins was selected for biomarker testing using an independent cohort of plasma and serum samples from well-diagnosed patients with pancreatic cancer, chronic pancreatitis, and nonpancreatic disease controls. Using ELISA methodology, the performance of each of these protein candidates was benchmarked against CA19-9, the current gold standard for a pancreatic cancer blood test. A composite marker of TIMP1 and ICAM1 demonstrate significantly better performance than CA19-9 in distinguishing pancreatic cancer from the nonpancreatic disease controls and chronic pancreatitis controls. In addition, protein AZGP1 was identified as a biomarker candidate for chronic pancreatitis. The discovery and technical challenges associated with plasma-based quantitative proteomics are discussed and may benefit the development of plasma proteomics technology in general. The protein candidates identified in this study provide a biomarker candidate pool for future investigations.
Collapse
Affiliation(s)
- Sheng Pan
- Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Quiros-Gonzalez I, Sainz RM, Hevia D, Mayo JC. MnSOD drives neuroendocrine differentiation, androgen independence, and cell survival in prostate cancer cells. Free Radic Biol Med 2011; 50:525-36. [PMID: 21056653 DOI: 10.1016/j.freeradbiomed.2010.10.715] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/19/2010] [Accepted: 10/28/2010] [Indexed: 12/14/2022]
Abstract
An increase in neuroendocrine (NE) cell number has been associated with progression of prostate tumor, one of the most frequent cancers among Western males. We previously reported that mitochondrial manganese superoxide dismutase (MnSOD) increases during the NE differentiation process. The goal of this study was to find whether MnSOD up-regulation is enough to induce NE differentiation. Several human prostate cancer LNCaP cell clones stably overexpressing MnSOD were characterized and two were selected (MnSOD-S4 and MnSOD-S12). MnSOD overexpression induces NE morphological features as well as coexpression of the NE marker synaptophysin. Both MnSOD clones exhibit lower superoxide levels and higher H(2)O(2) levels. MnSOD-overexpressing cells show higher proliferation rates in complete medium, but in steroid-free medium MnSOD-S12 cells are still capable of proliferation. MnSOD up-regulation decreases androgen receptor and prevents its nuclear translocation. MnSOD also induces up-regulation of Bcl-2 and prevents docetaxel-, etoposide-, or TNF-induced cell death. Finally, MnSOD-overexpressing cells enhance growth of androgen-independent PC-3 cells but reduce growth of androgen-dependent cells. These results indicate that redox modulation caused by MnSOD overexpression explains most NE-like features, including morphological changes, NE marker expression, androgen independence, inhibition of apoptosis, and enhancement of cell growth. Many of these events can be associated with the androgen dependent-independent transition during prostate cancer progression.
Collapse
Affiliation(s)
- Isabel Quiros-Gonzalez
- Instituto Universitario de Oncología del Principado de Asturias, Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | | | | | | |
Collapse
|
39
|
Chang KP, Wu CC, Chen HC, Chen SJ, Peng PH, Tsang NM, Lee LY, Liu SC, Liang Y, Lee YS, Hao SP, Chang YS, Yu JS. Identification of candidate nasopharyngeal carcinoma serum biomarkers by cancer cell secretome and tissue transcriptome analysis: potential usage of cystatin A for predicting nodal stage and poor prognosis. Proteomics 2010; 10:2644-60. [PMID: 20461718 DOI: 10.1002/pmic.200900620] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is usually diagnosed at advanced clinical stages, resulting in poor outcomes. To discover serum biomarkers for improved NPC diagnosis and/or management, we simultaneously analyzed the NPC cell secretome and tissue transcriptome to identify candidate genes/proteins that are highly upregulated in NPC tissues and also secreted/released from NPC cells. Among the 30 candidates identified, 11 proteins were chosen for further validation using the serum samples from NPC patients and healthy controls, including cystatin A, cathepsin B, manganese superoxide dismutase and matrix metalloproteinase 2. The results showed that serum levels of all the four proteins were indeed higher in NPC patients versus healthy controls and that the use of a three-marker panel (cystatin A, manganese superoxide dismutase and matrix metalloproteinase 2) can contribute to a better NPC detection than each marker alone. In addition, a higher pretreated serum level of cystatin A was found to be associated with a higher nodal stage and poorer prognosis of NPC patients and cystatin A could modulate the migration and invasion of NPC cells in vitro. Altogether, our results indicate that analysis of both the cancer cell secretome and tissue transcriptome is a feasible strategy for efficient identification of novel NPC serum marker panel.
Collapse
Affiliation(s)
- Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Elevated rates of reactive oxygen species (ROS) have been detected in almost all cancers, where they promote many aspects of tumour development and progression. However, tumour cells also express increased levels of antioxidant proteins to detoxify from ROS, suggesting that a delicate balance of intracellular ROS levels is required for cancer cell function. Further, the radical generated, the location of its generation, as well as the local concentration is important for the cellular functions of ROS in cancer. A challenge for novel therapeutic strategies will be the fine tuning of intracellular ROS signalling to effectively deprive cells from ROS-induced tumour promoting events, towards tipping the balance to ROS-induced apoptotic signalling. Alternatively, therapeutic antioxidants may prevent early events in tumour development, where ROS are important. However, to effectively target cancer cells specific ROS-sensing signalling pathways that mediate the diverse stress-regulated cellular functions need to be identified. This review discusses the generation of ROS within tumour cells, their detoxification, their cellular effects, as well as the major signalling cascades they utilize, but also provides an outlook on their modulation in therapeutics.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville FL 32224, USA
| | | |
Collapse
|
41
|
Kim SC, Magesh V, Jeong SJ, Lee HJ, Ahn KS, Lee HJ, Lee EO, Kim SH, Lee MH, Kim JH, Kim SH. Ethanol extract of Ocimum sanctum exerts anti-metastatic activity through inactivation of matrix metalloproteinase-9 and enhancement of anti-oxidant enzymes. Food Chem Toxicol 2010; 48:1478-82. [PMID: 20233602 DOI: 10.1016/j.fct.2010.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 02/11/2010] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Ocimum sanctum has been known to possess various beneficial properties including anti-oxidative, anti-inflammatory and anti-cancer activities. In the present study, we investigated that ethanol extracts of O. sanctum (EEOS) had anti-metastatic activity through activation of anti-oxidative enzymes. EEOS exerted cytotoxicity against Lewis lung carcinoma (LLC) cells. Also, EEOS significantly inhibited cell adhesion and invasion as well as activities of matrix metalloproteinase-9 (MMP-9), but not MMP-2, indicating the important role of MMP-9 in anti-metastatic regulation of EEOS. In addition, EEOS significantly reduced the tumor nodule formation and lung weight in LLC-injected mice. Inhibitory effect of EEOS on metastasis was further confirmed by using hematoxylin and eosin (H&E) staining. Notably, we also found that EEOS enhanced activities of anti-oxidative enzymes such as superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) in a concentration-dependent manner. Taken together, our findings support that EEOS can be a potent anti-metastatic candidate through inactivation of MMP-9 and enhancement of anti-oxidant enzymes.
Collapse
Affiliation(s)
- Sun-Chae Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Measurement of superoxide dismutase, catalase and glutathione peroxidase in cultured cells and tissue. Nat Protoc 2009; 5:51-66. [PMID: 20057381 DOI: 10.1038/nprot.2009.197] [Citation(s) in RCA: 835] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Cells contain a large number of antioxidants to prevent or repair the damage caused by reactive oxygen species, as well as to regulate redox-sensitive signaling pathways. General protocols are described to measure the antioxidant enzyme activity of superoxide dismutase (SOD), catalase and glutathione peroxidase. The SODs convert superoxide radical into hydrogen peroxide and molecular oxygen, whereas the catalase and peroxidases convert hydrogen peroxide into water. In this way, two toxic species, superoxide radical and hydrogen peroxide, are converted to the harmless product water. Western blots, activity gels and activity assays are various methods used to determine protein and activity in both cells and tissue depending on the amount of protein required for each assay. Other techniques including immunohistochemistry and immunogold can further evaluate the levels of the various antioxidant enzymes in tissues and cells. In general, these assays require 24-48 h to complete.
Collapse
|
43
|
Nolan KA, Doncaster JR, Dunstan MS, Scott KA, Frenkel AD, Siegel D, Ross D, Barnes J, Levy C, Leys D, Whitehead RC, Stratford IJ, Bryce RA. Synthesis and Biological Evaluation of Coumarin-Based Inhibitors of NAD(P)H: Quinone Oxidoreductase-1 (NQO1). J Med Chem 2009; 52:7142-56. [PMID: 19877692 DOI: 10.1021/jm9011609] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | - David Siegel
- Department of Pharmaceutical Sciences, University of Colorado Denver School of Pharmacy, Aurora, Colorado
| | - David Ross
- Department of Pharmaceutical Sciences, University of Colorado Denver School of Pharmacy, Aurora, Colorado
| | | | | | | | | | | | | |
Collapse
|
44
|
Bharadwaj U, Li M, Chen C, Yao Q. Mesothelin-induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol Cancer Res 2009; 6:1755-65. [PMID: 19010822 DOI: 10.1158/1541-7786.mcr-08-0095] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesothelin (MSLN) is a cell surface glycoprotein that is overexpressed in human pancreatic cancer. Although its value as a tumor marker for diagnosis and prognosis and as a preferred target of immunointervention has been evaluated, there is little information on the growth advantage of MSLN on tumor cells. In this study, we examined the effect of MSLN on pancreatic cancer cell proliferation, cell cycle progression, expression of cell cycle regulatory proteins, and signal transduction pathways in two pancreatic cancer cell lines, MIA-MSLN (overexpressing MSLN in MIA PaCa-2 cells) and BxPC-siMSLN (silencing MSLN in BxPC-3 cells). Increased cyclin E and cyclin-dependent kinase 2 expression found in MIA-MSLN cells correlated with significantly increased cell proliferation and faster cell cycle progression compared with control cells. BxPC-siMSLN cells showed slower proliferation and slower entry into the S phase than control cells. Signal transducer and activator of transcription protein 3 (Stat3) was constitutively activated in MIA-MSLN cells, but not in control cells. Inhibition of Stat3 activation in MIA-MSLN cells by the Janus-activated kinase-selective inhibitor tyrphostin AG490 was followed by a marked decrease in proliferation of the cells. Small interfering RNA against Stat3 significantly reduced the MIA-MSLN cell cycle progression with a concomitant decrease in cyclin E expression. Our data indicate that overexpression of MSLN in pancreatic cancer cells leads to constitutive activation of the transcription factor Stat3, which results in enhanced expression of cyclin E and cyclin E/cyclin-dependent kinase 2 complex formation as well as increased G(1)-S transition.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Molecular Surgeon Research Center, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
45
|
Hempel N, Ye H, Abessi B, Mian B, Melendez JA. Altered redox status accompanies progression to metastatic human bladder cancer. Free Radic Biol Med 2009; 46:42-50. [PMID: 18930813 PMCID: PMC2630461 DOI: 10.1016/j.freeradbiomed.2008.09.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 09/04/2008] [Accepted: 09/15/2008] [Indexed: 02/04/2023]
Abstract
The role of reactive oxygen species (ROS) in bladder cancer progression remains an unexplored field. Expression levels of enzymes regulating ROS levels are often altered in cancer. A search of publicly available microarray data reveals that expression of mitochondrial manganese superoxide dismutase (Sod2), responsible for the conversion of superoxide (O(2)(-)) to hydrogen peroxide (H(2)O(2)), is consistently increased in high-grade and advanced-stage bladder tumors. We aimed to identify the role of Sod2 expression and ROS in bladder cancer. Using an in vitro human bladder tumor model we monitored the redox state of both nonmetastatic (253J) and highly metastatic (253J B-V) bladder tumor cell lines. 253J B-V cells displayed significantly higher Sod2 protein and activity levels compared to their parental 253J cell line. The increase in Sod2 expression was accompanied by a significant decrease in catalase activity, resulting in a net increase in H(2)O(2) production in the 253J B-V cell line. Expression of the prometastatic and proangiogenic factors matrix metalloproteinase 9 (MMP-9) and vascular endothelial-derived growth factor (VEGF), respectively, was upregulated in the metastatic line. Expression of both MMP-9 and VEGF was shown to be H(2)O(2)-dependent, as removal of H(2)O(2) by overexpression of catalase attenuated their expression. Similarly, expression of catalase effectively reduced the clonogenic activity of 253J B-V cells. These findings indicate that metastatic bladder cancer cells display an altered antioxidant expression profile, resulting in a net increase in ROS production, which leads to the induction of redox-sensitive protumorigenic and prometastatic genes such as VEGF and MMP-9.
Collapse
Affiliation(s)
- Nadine Hempel
- Center for Immunology and Microbial Disease, Albany Medical College, NY 12208, USA
| | | | | | | | | |
Collapse
|
46
|
Hitchler MJ, Oberley LW, Domann FE. Epigenetic silencing of SOD2 by histone modifications in human breast cancer cells. Free Radic Biol Med 2008; 45:1573-80. [PMID: 18845242 PMCID: PMC2633123 DOI: 10.1016/j.freeradbiomed.2008.09.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 09/04/2008] [Accepted: 09/07/2008] [Indexed: 11/27/2022]
Abstract
Many breast cancer cells typically exhibit lower expression of manganese superoxide dismutase (MnSOD) compared to the normal cells from which they arise. This decrease can often be attributed to a defect in the transcription of SOD2, the gene encoding MnSOD; however, the mechanism responsible for this change remains unclear. Here, we describe how altered histone modifications and a repressive chromatin structure constitute an epigenetic process to down regulate SOD2 in human breast carcinoma cell lines. Utilizing chromatin immunoprecipitation (ChIP) we observed decreased levels of dimethyl H3K4 and acetylated H3K9 at key regulatory elements of the SOD2 gene. Consistent with these results, we show that loss of these histone modifications creates a repressive chromatin structure at SOD2. Transcription factor ChIP experiments revealed that this repressive chromatin structure influences the binding of SP-1, AP-1, and NFkappaB to SOD2 regulatory cis-elements in vivo. Lastly, we show that treatment with the histone deacetylase inhibitors trichostatin A and sodium butyrate can reactivate SOD2 expression in breast cancer cell lines. Taken together, these results indicate that epigenetic silencing of SOD2 could be facilitated by changes in histone modifications and represent one mechanism leading to the altered expression of MnSOD observed in many breast cancers.
Collapse
Affiliation(s)
- Michael J Hitchler
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
47
|
Teoh MLT, Sun W, Smith BJ, Oberley LW, Cullen JJ. Modulation of reactive oxygen species in pancreatic cancer. Clin Cancer Res 2008; 13:7441-50. [PMID: 18094428 DOI: 10.1158/1078-0432.ccr-07-0851] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of the present study was to compare the effects of the three different forms of the antioxidant enzyme superoxide dismutase [i.e., manganese superoxide dismutase (MnSOD), copper zinc superoxide dismutase (CuZnSOD), and extracellular superoxide dismutase (EcSOD)] on the malignant phenotype of human pancreatic cancer. EXPERIMENTAL DESIGN Human pancreatic cancer cell lines were infected with adenoviral vectors containing the cDNAs for three different forms of the antioxidant enzyme SOD. Intratumoral injections of the adenoviral vectors were used in nude mice with human tumor xenografts. RESULTS Increases in immunoreactive protein and enzymatic activity were seen after infections with the AdMnSOD, AdCuZnSOD, or AdEcSOD constructs. Increased SOD activity decreased superoxide levels and increased hydrogen peroxide levels. Increasing SOD levels correlated with increased doubling time. Cell growth and plating efficiency decreased with increasing amounts of the adenoviral constructs, with the AdCuZnSOD vector having the greatest effect in decreasing in vitro tumor growth. In contrast, inhibiting endogenous SOD with small interfering RNA increased superoxide levels and promoted tumor growth. Of the three SODs, tumors grew the slowest and survival was increased the greatest in nude mice injected with the AdEcSOD construct. CONCLUSIONS Scavenging plasma membrane-generated superoxide may prove beneficial for suppression of pancreatic cancer growth.
Collapse
Affiliation(s)
- Melissa L T Teoh
- Department of Surgery and Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
48
|
Nolan KA, Zhao H, Faulder PF, Frenkel AD, Timson DJ, Siegel D, Ross D, Burke Jr. TR, Stratford IJ, Bryce RA. Coumarin-Based Inhibitors of Human NAD(P)H:Quinone Oxidoreductase-1. Identification, Structure–Activity, Off-Target Effects and In Vitro Human Pancreatic Cancer Toxicity. J Med Chem 2007; 50:6316-25. [DOI: 10.1021/jm070472p] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Karen A. Nolan
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - He Zhao
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - Paul F. Faulder
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - A. David Frenkel
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - David J. Timson
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - David Siegel
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - David Ross
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - Terrence R. Burke Jr.
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - Ian J. Stratford
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| | - Richard A. Bryce
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, U.K., Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, U.K., and Department of Pharmaceutical Sciences and Cancer Center, School of Pharmacy, University of Colorado and Health Sciences Center, Denver, Colorado 80220
| |
Collapse
|
49
|
Mohr A, Büneker C, Gough RP, Zwacka RM. MnSOD protects colorectal cancer cells from TRAIL-induced apoptosis by inhibition of Smac/DIABLO release. Oncogene 2007; 27:763-74. [PMID: 17653087 DOI: 10.1038/sj.onc.1210673] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mitochondrial enzyme manganese superoxide dismutase (MnSOD) has been shown to have two faces with regard to its role in tumor development. On the one side, it is well documented that overexpression of MnSOD slows down cancer cell growth, whereas on the other side MnSOD also has a metastasis-promoting activity. We set out to examine the role of MnSOD in tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, thought to be a first-line tumor surveillance mechanism and failure to undergo apoptosis might contribute to metastasis formation. We show that overexpression of MnSOD at moderate levels is able to protect cells from TRAIL-induced apoptosis. While caspase-8 activation and Bid cleavage were not affected by MnSOD, we detected a marked decrease in caspase-3 activation pointing to a mitochondrial resistance mechanism. Indeed, we found that MnSOD-overexpressing cells showed reduced cytochrome c and no Smac/DIABLO release into the cytosol. The resulting lack of X-linked inhibitor of apoptosis (XIAP) inhibition by cytosolic Smac/DIABLO most likely caused the TRAIL resistance as RNAi against XIAP-rescued caspase-3 activity and TRAIL sensitivity. Our results show that reactive oxygen species are involved in TRAIL-induced Smac/DIABLO release and in TRAIL-triggered apoptosis. Hence, high levels of MnSOD, which decompose and neutralize these reactive oxygen species, might contribute to metastasis formation by allowing disseminated tumor cells to escape from TRAIL-mediated tumor surveillance. As part of TRAIL regimens, adjuvant treatment with XIAP inhibitors in the form of Smac/DIABLO mimetics or MnSOD inhibitors might be able to break TRAIL resistance of malignant tumor cells.
Collapse
Affiliation(s)
- A Mohr
- National Centre for Biomedical Engineering Science, Molecular Therapeutics Group, National University of Ireland Galway, Galway, Ireland
| | | | | | | |
Collapse
|