51
|
Nguyen H, Syed V. Progesterone inhibits growth and induces apoptosis in cancer cells through modulation of reactive oxygen species. Gynecol Endocrinol 2011; 27:830-6. [PMID: 21171931 DOI: 10.3109/09513590.2010.538100] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Progesterone (P4) has been implicated as a protective factor for ovarian and endometrial cancers, yet little is known about its mechanism of action. We have shown apoptosis in ovarian and endometrial cancer cells with high doses of P4. Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells. The goal of this study was to assess the effect of P4 on cell growth, ROS generation, oxidative stress markers, and the expression of antioxidant proteins. METHODS All experiments were performed in vitro using cancer cell lines. Cell proliferation was determined using MTS proliferation assay. Production of ROS in cells was measured with the ROS indicator dye, aminophenyl fluorescein. Alterations in expression of antioxidant and apoptotic proteins were assessed by Western blotting. RESULTS The exposure of ovarian and endometrial cancer cell cultures to various doses of P4 caused a dose-dependent decrease in cell viability and the activation of caspase-3. Levels of ROS, markers of oxidative stress, and antioxidant proteins were elevated in cancer cells compared to normal cells and a marked decrease in their expression was seen following P4 treatment. In cancer cells, ROS was elevated while p-53 expression was low. P4 exposure of cells resulted in increased p-53 and BAX and decreased BCL-2 expression. CONCLUSIONS The data indicates that P4 has antioxidant effects. It alleviates ROS stress and causes apoptosis by upregulating proapoptotic (p-53 and BAX) and decreasing antiapoptotic (BCL-2) gene expression in cancer cells. These findings could have potential therapeutic implications.
Collapse
Affiliation(s)
- Huyen Nguyen
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814-4799, USA
| | | |
Collapse
|
52
|
Das UN. Essential fatty acids enhance free radical generation and lipid peroxidation to induce apoptosis of tumor cells. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/clp.11.34] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
53
|
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: 124] [Impact Index Per Article: 8.9] [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
|
54
|
Tsai S, Hou M, Wu S, Hu B, Yang S, Chen W, Chai C, Ma H, Tsai L. Expression of manganese superoxide dismutase in patients with breast cancer. Kaohsiung J Med Sci 2011; 27:167-72. [PMID: 21527182 PMCID: PMC11916061 DOI: 10.1016/j.kjms.2010.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 11/26/2010] [Indexed: 12/26/2022] Open
Abstract
Breast cancer has become the second leading cancer among females in Taiwan. Even though the etiology of breast cancer is multifactorial, oxidative stress plays an important role in the carcinogenesis. The purpose of this study was to investigate the expression of manganese superoxide dismutase (MnSOD), one of the major antioxidant enzymes that is involved against oxidative stress, in adjacent cancer-free breast tissues and neoplasm tissues within the same patient. Sixty-five breast cancer patients' formalin-fixed tissue blocks, including ductal carcinoma in situ (DCIS) tissues, invasive ductal carcinoma (IDC) tissues, and adjacent cancer-free tissues, were evaluated by immunohistochemical stain. Meanwhile, their demographic and clinical information was also collected. The combined scores of MnSOD-positive cell proportion and MnSOD staining intensity were compared for different tissues within the same patient. The results showed that the mean combined scores of MnSOD expression in adjacent cancer-free tissues (6.33), IDC (5.30), and DCIS (3.78) were significantly different when assessed by repeated-measurement analysis of variance (F=14.17, p<0.001). Additionally, the results revealed that the distribution of strong MnSOD protein expression was 80.0%, 72.3%, and 52.3% in adjacent cancer-free tissues, IDC, and DCIS, respectively. However, there was no statistically significant relationship between the expression of MnSOD and grades of breast cancer or other clinicopathologic variables. We suggest that the expression of MnSOD in neoplasm tissues, independent of the clinicopathologic characters, plays a critical role in breast cancer biology.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Female
- Humans
- Middle Aged
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Superoxide Dismutase/metabolism
- Tumor Burden
Collapse
Affiliation(s)
- Shih‐Meng Tsai
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming‐Feng Hou
- Department of Surgery, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Chung‐Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Szu‐Hsien Wu
- Division of Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang‐Ming University, Taipei, Taiwan
| | - Bao‐Wen Hu
- Faculty of Medical Laboratory and Biotechnology Science, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheau‐Fang Yang
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan‐Tzu Chen
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee‐Yin Chai
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsu Ma
- Division of Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang‐Ming University, Taipei, Taiwan
| | - Li‐Yu Tsai
- Faculty of Medical Laboratory and Biotechnology Science, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
55
|
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: 3.0] [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
|
56
|
Hillegass JM, Shukla A, MacPherson MB, Lathrop SA, Alexeeva V, Perkins TN, van der Vliet A, Vacek PM, Gunter ME, Mossman BT. Mechanisms of oxidative stress and alterations in gene expression by Libby six-mix in human mesothelial cells. Part Fibre Toxicol 2010; 7:26. [PMID: 20831825 PMCID: PMC2945990 DOI: 10.1186/1743-8977-7-26] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 09/11/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Exposures to an amphibole fiber in Libby, Montana cause increases in malignant mesothelioma (MM), a tumor of the pleural and peritoneal cavities with a poor prognosis. Affymetrix microarray/GeneSifter analysis was used to determine alterations in gene expression of a human mesothelial cell line (LP9/TERT-1) by a non-toxic concentration (15×10(6) μm2/cm2) of unprocessed Libby six-mix and negative (glass beads) and positive (crocidolite asbestos) controls. Because manganese superoxide dismutase (MnSOD; SOD2) was the only gene upregulated significantly (p < 0.05) at both 8 and 24 h, we measured SOD protein and activity, oxidative stress and glutathione (GSH) levels to better understand oxidative events after exposure to non-toxic (15×10(6) μm2/cm2) and toxic concentrations (75×10(6) μm2/cm2) of Libby six-mix. RESULTS Exposure to 15×10(6) μm2/cm2 Libby six-mix elicited significant (p < 0.05) upregulation of one gene (SOD2; 4-fold) at 8 h and 111 gene changes at 24 h, including a 5-fold increase in SOD2. Increased levels of SOD2 mRNA at 24 h were also confirmed in HKNM-2 normal human pleural mesothelial cells by qRT-PCR. SOD2 protein levels were increased at toxic concentrations (75×10(6) μm2/cm2) of Libby six-mix at 24 h. In addition, levels of copper-zinc superoxide dismutase (Cu/ZnSOD; SOD1) protein were increased at 24 h in all mineral groups. A dose-related increase in SOD2 activity was observed, although total SOD activity remained unchanged. Dichlorodihydrofluorescein diacetate (DCFDA) fluorescence staining and flow cytometry revealed a dose- and time-dependent increase in reactive oxygen species (ROS) production by LP9/TERT-1 cells exposed to Libby six-mix. Both Libby six-mix and crocidolite asbestos at 75×10(6) μm2/cm2 caused transient decreases (p < 0.05) in GSH for up to 24 h and increases in gene expression of heme oxygenase 1 (HO-1) in LP9/TERT-1 and HKNM-2 cells. CONCLUSIONS Libby six-mix causes multiple gene expression changes in LP9/TERT-1 human mesothelial cells, as well as increases in SOD2, increased production of oxidants, and transient decreases in intracellular GSH. These events are not observed at equal surface area concentrations of nontoxic glass beads. Results support a mechanistic basis for the importance of SOD2 in proliferation and apoptosis of mesothelial cells and its potential use as a biomarker of early responses to mesotheliomagenic minerals.
Collapse
Affiliation(s)
- Jedd M Hillegass
- Department of Pathology, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Deregulation of manganese superoxide dismutase (SOD2) expression and lymph node metastasis in tongue squamous cell carcinoma. BMC Cancer 2010; 10:365. [PMID: 20618948 PMCID: PMC2911422 DOI: 10.1186/1471-2407-10-365] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 07/09/2010] [Indexed: 01/25/2023] Open
Abstract
Background Lymph node metastasis is a critical event in the progression of tongue squamous cell carcinoma (TSCC). The identification of biomarkers associated with the metastatic process would provide critical prognostic information to facilitate clinical decision making. Previous studies showed that deregulation of manganese superoxide dismutase (SOD2) expression is a frequent event in TSCC and may be associated with enhanced cell invasion. The purpose of this study is to further evaluate whether the expression level of SOD2 is correlated with the metastatic status in TSCC patients. Methods We first examined the SOD2 expression at mRNA level on 53 TSCC and 22 normal control samples based on pooled-analysis of existing microarray datasets. To confirm our observations, we examined the expression of SOD2 at protein level on an additional TSCC patient cohort (n = 100), as well as 31 premalignant dysplasias, 15 normal tongue mucosa, and 32 lymph node metastatic diseases by immunohistochemistry (IHC). Results The SOD2 mRNA level in primary TSCC tissue is reversely correlated with lymph node metastasis in the first TSCC patient cohort. The SOD2 protein level in primary TSCC tissue is also reversely correlated with lymph node metastasis in the second TSCC patient cohort. Deregulation of SOD2 expression is a common event in TSCC and appears to be associated with disease progression. Statistical analysis revealed that the reduced SOD2 expression in primary tumor tissue is associated with lymph node metastasis in both TSCC patient cohorts examined. Conclusions Our study suggested that the deregulation of SOD2 in TSCC has potential predictive values for lymph node metastasis, and may serve as a therapeutic target for patients at risk of metastasis.
Collapse
|
58
|
McAtee BL, Yager JD. Manganese superoxide dismutase: effect of the ala16val polymorphism on protein, activity, and mRNA levels in human breast cancer cell lines and stably transfected mouse embryonic fibroblasts. Mol Cell Biochem 2009; 335:107-18. [PMID: 19756960 DOI: 10.1007/s11010-009-0247-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 09/02/2009] [Indexed: 11/30/2022]
Abstract
The manganese superoxide dismutase (MnSOD) ala16val polymorphism has been associated with various diseases including breast cancer. In the present study, we investigated levels of MnSOD protein, enzymatic activity, and mRNA with respect to MnSOD genotype in several human breast carcinoma cell lines and in mouse embryonic fibroblasts (MEF), developed from the MnSOD knockout mouse, stably expressing human MnSOD-ala and MnSOD-val. In human breast cell lines, the MnSOD-ala allele was associated with increased levels of MnSOD protein and MnSOD protein per unit mRNA. In the MEF transformants, MnSOD activity correlated fairly well with MnSOD protein levels. MnSOD mRNA expression was significantly lower in MnSOD-ala versus MnSOD-val lines. MnSOD protein and activity levels were not related to MnSOD genotype in the transformed MEF, although, as observed in the human breast cell lines, the MEF human MnSOD-ala lines produced significantly more human MnSOD protein per unit mRNA than the human MnSOD-val lines. This suggests that there is more efficient production of MnSOD-ala protein compared to MnSOD-val protein. Examination of several indicators of reactive oxygen species levels, including superoxide and hydrogen peroxide, in wild-type MEF and in MEF expressing similar elevated amounts of MnSOD-ala or val activity did not show differences related to the levels of MnSOD protein expression. In conclusion, in both human breast carcinoma cell lines and MEF cell lines stably transfected with human MnSOD, the MnSOD-ala allele was associated with increased production of MnSOD protein per unit mRNA indicating a possible imbalance in MnSOD protein production from the MnSOD-val mRNA.
Collapse
Affiliation(s)
- Britt L McAtee
- Division of Toxicology, Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | | |
Collapse
|
59
|
Hempel N, Ye H, Abessia 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: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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, Albany, NY 12008
| | - Hanqing Ye
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12008
| | - Bryan Abessia
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12008
| | - Badar Mian
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12008
- Department of Urology, Albany Medical College, Albany, NY 12208
| | - J. Andres Melendez
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12008
| |
Collapse
|
60
|
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: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
61
|
Reddy SP, Britto R, Vinnakota K, Aparna H, Sreepathi HK, Thota B, Kumari A, Shilpa BM, Vrinda M, Umesh S, Samuel C, Shetty M, Tandon A, Pandey P, Hegde S, Hegde AS, Balasubramaniam A, Chandramouli BA, Santosh V, Kondaiah P, Somasundaram K, Rao MRS. Novel glioblastoma markers with diagnostic and prognostic value identified through transcriptome analysis. Clin Cancer Res 2008; 14:2978-87. [PMID: 18483363 DOI: 10.1158/1078-0432.ccr-07-4821] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Current methods of classification of astrocytoma based on histopathologic methods are often subjective and less accurate. Although patients with glioblastoma have grave prognosis, significant variability in patient outcome is observed. Therefore, the aim of this study was to identify glioblastoma diagnostic and prognostic markers through microarray analysis. EXPERIMENTAL DESIGN We carried out transcriptome analysis of 25 diffusely infiltrating astrocytoma samples [WHO grade II--diffuse astrocytoma, grade III--anaplastic astrocytoma, and grade IV--glioblastoma (GBM)] using cDNA microarrays containing 18,981 genes. Several of the markers identified were also validated by real-time reverse transcription quantitative PCR and immunohistochemical analysis on an independent set of tumor samples (n = 100). Survival analysis was carried out for two markers on another independent set of retrospective cases (n = 51). RESULTS We identified several differentially regulated grade-specific genes. Independent validation by real-time reverse transcription quantitative PCR analysis found growth arrest and DNA-damage-inducible alpha (GADD45alpha) and follistatin-like 1 (FSTL1) to be up-regulated in most GBMs (both primary and secondary), whereas superoxide dismutase 2 and adipocyte enhancer binding protein 1 were up-regulated in the majority of primary GBM. Further, identification of the grade-specific expression of GADD45alpha and FSTL1 by immunohistochemical staining reinforced our findings. Analysis of retrospective GBM cases with known survival data revealed that cytoplasmic overexpression of GADD45alpha conferred better survival while the coexpression of FSTL1 with p53 was associated with poor survival. CONCLUSIONS Our study reveals that GADD45alpha and FSTLI are GBM-specific whereas superoxide dismutase 2 and adipocyte enhancer binding protein 1 are primary GBM-specific diagnostic markers. Whereas GADD45alpha overexpression confers a favorable prognosis, FSTL1 overexpression is a hallmark of poor prognosis in GBM patients.
Collapse
Affiliation(s)
- Sreekanth P Reddy
- Department of Molecular Reproduction, Indian Institute of Science, Bangalore, Karnataka, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Ye H, Yu T, Temam S, Ziober BL, Wang J, Schwartz JL, Mao L, Wong DT, Zhou X. Transcriptomic dissection of tongue squamous cell carcinoma. BMC Genomics 2008; 9:69. [PMID: 18254958 PMCID: PMC2262071 DOI: 10.1186/1471-2164-9-69] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Accepted: 02/06/2008] [Indexed: 11/12/2022] Open
Abstract
Background The head and neck/oral squamous cell carcinoma (HNOSCC) is a diverse group of cancers, which develop from many different anatomic sites and are associated with different risk factors and genetic characteristics. The oral tongue squamous cell carcinoma (OTSCC) is one of the most common types of HNOSCC. It is significantly more aggressive than other forms of HNOSCC, in terms of local invasion and spread. In this study, we aim to identify specific transcriptomic signatures that associated with OTSCC. Results Genome-wide transcriptomic profiles were obtained for 53 primary OTSCCs and 22 matching normal tissues. Genes that exhibit statistically significant differences in expression between OTSCCs and normal were identified. These include up-regulated genes (MMP1, MMP10, MMP3, MMP12, PTHLH, INHBA, LAMC2, IL8, KRT17, COL1A2, IFI6, ISG15, PLAU, GREM1, MMP9, IFI44, CXCL1), and down-regulated genes (KRT4, MAL, CRNN, SCEL, CRISP3, SPINK5, CLCA4, ADH1B, P11, TGM3, RHCG, PPP1R3C, CEACAM7, HPGD, CFD, ABCA8, CLU, CYP3A5). The expressional difference of IL8 and MMP9 were further validated by real-time quantitative RT-PCR and immunohistochemistry. The Gene Ontology analysis suggested a number of altered biological processes in OTSCCs, including enhancements in phosphate transport, collagen catabolism, I-kappaB kinase/NF-kappaB signaling cascade, extracellular matrix organization and biogenesis, chemotaxis, as well as suppressions of superoxide release, hydrogen peroxide metabolism, cellular response to hydrogen peroxide, keratinization, and keratinocyte differentiation in OTSCCs. Conclusion In summary, our study provided a transcriptomic signature for OTSCC that may lead to a diagnosis or screen tool and provide the foundation for further functional validation of these specific candidate genes for OTSCC.
Collapse
Affiliation(s)
- Hui Ye
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Up-regulated manganese superoxide dismutase expression increases apoptosis resistance in human esophageal squamous cell carcinomas. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200712010-00006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
64
|
The “One Source” cohort — evaluating the suitability of the human toenail as a manganese biomonitor. J Radioanal Nucl Chem 2007. [DOI: 10.1007/s10967-007-0407-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
65
|
Chuang TC, Liu JY, Lin CT, Tang YT, Yeh MH, Chang SC, Li JW, Kao MC. Human manganese superoxide dismutase suppresses HER2/neu-mediated breast cancer malignancy. FEBS Lett 2007; 581:4443-9. [PMID: 17719580 DOI: 10.1016/j.febslet.2007.08.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 08/09/2007] [Accepted: 08/09/2007] [Indexed: 10/22/2022]
Abstract
The up-regulation of HER2/neu is associated with human malignancies and is a useful target for developing anticancer drugs. Overexpression of human manganese superoxide dismutase (MnSOD) has been demonstrated to effectively suppress various carcinoma cells, including breast carcinomas, in vitro and in vivo. This study demonstrates that MnSOD effectively suppresses HER2/neu oncogene expression at the transcriptional level. Additionally, stable transfection was used and the MnSOD-transfected human breast cancer clones were found to be able to down-regulate the endogenous production of p185(HER2/neu). Furthermore, the MnSOD-overexpressing stable transfectants exhibited reduced soft-agarose colony-forming ability and metastatic properties, unlike control cell lines. These data suggest that MnSOD may be useful in treating HER2/neu-mediated human breast tumor malignancy.
Collapse
Affiliation(s)
- Tzu-Chao Chuang
- Department of Chemistry, Tamkang University, Tamsui, Taiwan, ROC.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
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: 2.2] [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
|
67
|
Hwang IT, Chung YM, Kim JJ, Chung JS, Kim BS, Kim HJ, Kim JS, Yoo YD. Drug resistance to 5-FU linked to reactive oxygen species modulator 1. Biochem Biophys Res Commun 2007; 359:304-10. [PMID: 17537404 DOI: 10.1016/j.bbrc.2007.05.088] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 05/14/2007] [Indexed: 11/29/2022]
Abstract
While acute oxidative stress triggers cell apoptosis or necrosis, persistent oxidative stress induces genomic instability and has been implicated in tumor progression and drug resistance. In a previous report, we demonstrated that reactive oxygen species modulator 1 (Romo1) expression was up-regulated in most cancer cell lines and suggested that increased Romo1 expression might confer chronic oxidative stress to tumor cells. In this study, we show that enforced Romo1 expression induces reactive oxygen species (ROS) production in the mitochondria leading to massive cell death. However, tumor cells that adapt to oxidative stress by increasing manganese superoxide dismutase (MnSOD), Prx I, and Bcl-2 showed drug resistance to 5-FU. To elucidate the relationship between 5-FU-induced ROS production and Romo1 expression, Romo1 siRNA was used to inhibit 5-FU-triggered Romo1 induction. Romo1 siRNA treatment efficiently blocked 5-FU-induced ROS generation, demonstrating that 5-FU treatment stimulated ROS production through Romo1 induction. Based on these results we suggest that cellular adaptive response to Romo1-induced ROS is another mechanism of drug resistance to 5-FU and Romo1 expression may provide a new clinical implication in drug resistance of cancer chemotherapy.
Collapse
Affiliation(s)
- In Tae Hwang
- Graduate School of Medicine, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Kattan Z, Minig V, Leroy P, Dauça M, Becuwe P. Role of manganese superoxide dismutase on growth and invasive properties of human estrogen-independent breast cancer cells. Breast Cancer Res Treat 2007; 108:203-15. [PMID: 17473980 DOI: 10.1007/s10549-007-9597-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Accepted: 04/05/2007] [Indexed: 10/23/2022]
Abstract
Manganese superoxide dismutase (MnSOD) is known to play a role in cancer. MnSOD exerts a tumor suppressive effect in estrogen-dependent human breast cancer cells. In the present study we investigated the in vitro role of MnSOD in the growth of some aggressive and highly metastatic estrogen-independent breast cancer cells, i.e., MDA-MB231 and SKBR3 cells. We show that estrogen-independent cells expressed a significantly higher basal MnSOD level compared to estrogen-dependent human breast cancer cell lines (MCF-7 and T47D). For MDA-MB231 cells, the high-MnSOD level was accompanied by an overproduction of intracellular hydrogen peroxide (H2O2) and by a low expression of the major H2O2-detoxifying enzymes, catalase, and peroxiredoxin 3, compared to MCF-7 cells. Suppression of MnSOD expression by antisense RNA was associated with a decrease of H2O2 content and caused a stimulation of growth with a reduced cell doubling time but induced a decrease of colony formation. Furthermore, treatment of MDA-MB231 cells with H2O2 scavengers markedly reduced tumor cell growth and colony formation. In addition, MnSOD suppression or treatment with H2O2 scavengers reduced the invasive properties of MDA-MB231 cells up to 43%, with a concomitant decrease of metalloproteinase-9 activity. We conclude that MnSOD plays a role in regulating tumor cell growth and invasive properties of estrogen-independent metastatic breast cancer cells. These action are mediated by MnSOD-dependent H2O2 production. In addition, these results suggest that MnSOD up-regulation may be one mechanism that contributes to the development of metastatic breast cancers.
Collapse
Affiliation(s)
- Zilal Kattan
- Laboratoire de Biologie Cellulaire du Développement, EA 3446-IFR111 (Proliférateurs de Peroxysomes), Université Henri Poincaré-Nancy I, Faculté des Sciences, B.P. 239, 54506, Vandoeure-les-Nancy, France
| | | | | | | | | |
Collapse
|
69
|
Karihtala P, Soini Y. Reactive oxygen species and antioxidant mechanisms in human tissues and their relation to malignancies. APMIS 2007; 115:81-103. [PMID: 17295675 DOI: 10.1111/j.1600-0463.2007.apm_514.x] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are formed in mammalian cells as a consequence of aerobic respiration. Despite multiple conserved redox modulating systems, a given proportion of ROS continuously escape from the mitochondrial respiratory chain, being sufficiently potent to damage cells in various ways, including numerous carcinogenic DNA mutations. Oxidative stress resulting from an imbalanced ratio between ROS production and detoxification may also disturb physiological signal transduction, lead to chain reactions in lipid layers, and damage DNA repair enzymes. The significance of ROS and antioxidant systems in carcinogenesis is still complicated and in many ways contradictory. Enhanced antioxidant mechanisms in tumor cells in vivo have been implicated in chemoresistance and lead to poor prognosis, whereas most in vitro studies have reported tumor-suppressing properties of antioxidant enzymes. The present review aims to clarify the significance of oxidative stress and the role of cell redox state modulating systems in human malignancies in light of the current literature.
Collapse
Affiliation(s)
- Peeter Karihtala
- Department of Pathology, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | | |
Collapse
|
70
|
Seifried HE, Anderson DE, Fisher EI, Milner JA. A review of the interaction among dietary antioxidants and reactive oxygen species. J Nutr Biochem 2007; 18:567-79. [PMID: 17360173 DOI: 10.1016/j.jnutbio.2006.10.007] [Citation(s) in RCA: 485] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 09/29/2006] [Accepted: 10/31/2006] [Indexed: 01/11/2023]
Abstract
During normal cellular activities, various processes inside of cells produce reactive oxygen species (ROS). Some of the most common ROS are hydrogen peroxide (H(2)O(2)), superoxide ion (O(2)(-)), and hydroxide radical (OH(-)). These compounds, when present in a high enough concentration, can damage cellular proteins and lipids or form DNA adducts that may promote carcinogenic activity. The purpose of antioxidants in a physiological setting is to prevent ROS concentrations from reaching a high-enough level within a cell that damage may occur. Cellular antioxidants may be enzymatic (catalase, glutathione peroxidase, superoxide dismutase) or nonenzymatic (glutathione, thiols, some vitamins and metals, or phytochemicals such as isoflavones, polyphenols, and flavanoids). Reactive oxygen species are a potential double-edged sword in disease prevention and promotion. Whereas generation of ROS once was viewed as detrimental to the overall health of the organism, advances in research have shown that ROS play crucial roles in normal physiological processes including response to growth factors, the immune response, and apoptotic elimination of damaged cells. Notwithstanding these beneficial functions, aberrant production or regulation of ROS activity has been demonstrated to contribute to the development of some prevalent diseases and conditions, including cancer and cardiovascular disease (CVD). The topic of antioxidant usage and ROS is currently receiving much attention because of studies linking the use of some antioxidants with increased mortality in primarily higher-risk populations and the lack of strong efficacy data for protection against cancer and heart disease, at least in populations with adequate baseline dietary consumption. In normal physiological processes, antioxidants effect signal transduction and regulation of proliferation and the immune response. Reactive oxygen species have been linked to cancer and CVD, and antioxidants have been considered promising therapy for prevention and treatment of these diseases, especially given the tantalizing links observed between diets high in fruits and vegetables (and presumably antioxidants) and decreased risks for cancer.
Collapse
Affiliation(s)
- Harold E Seifried
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20862, USA.
| | | | | | | |
Collapse
|
71
|
Abstract
A variety of cellular activities, including metabolism, growth, and death, are regulated and modulated by the redox status of the environment. A biphasic effect has been demonstrated on cellular proliferation with reactive oxygen species (ROS)-especially hydrogen peroxide and superoxide-in which low levels (usually submicromolar concentrations) induce growth but higher concentrations (usually >10-30 micromolar) induce apoptosis or necrosis. This phenomenon has been demonstrated for primary, immortalized and transformed cell types. However, the mechanism of the proliferative response to low levels of ROS is not well understood. Much of the work examining the signal transduction by ROS, including H(2)O(2), has been performed using doses in the lethal range. Although use of higher ROS doses have allowed the identification of important signal transduction pathways, these pathways may be activated by cells only in association with ROS-induced apoptosis and necrosis, and may not utilize the same pathways activated by lower doses of ROS associated with increased cell growth. Recent data has shown that low levels of exogenous H(2)O(2) up-regulate intracellular glutathione and activate the DNA binding activity toward antioxidant response element. The modulation of the cellular redox environment, through the regulation of cellular glutathione levels, may be a part of the hormetic effect shown by ROS on cell growth.
Collapse
Affiliation(s)
- Regina M Day
- Department of Pharmacology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814-4799, USA.
| | | |
Collapse
|
72
|
Lewis A, Du J, Liu J, Ritchie JM, Oberley LW, Cullen JJ. Metastatic progression of pancreatic cancer: changes in antioxidant enzymes and cell growth. Clin Exp Metastasis 2006; 22:523-32. [PMID: 16475022 DOI: 10.1007/s10585-005-4919-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 11/08/2005] [Indexed: 01/24/2023]
Abstract
Pancreatic cancer has a dismal prognosis due to the fact that patients present late when metastatic disease is already present. Previous studies have demonstrated that pancreatic cancer cells have decreased levels of MnSOD, which correlates well with increased rates of tumor cell proliferation. Recently, we have found that nude mice injected with MIA PaCa-2 human pancreatic cancer cells in the flank occasionally develop ascites and intra-abdominal metastatic deposits. Mice that developed ascites were sacrificed and the ascites cultured. Necropsy demonstrated metastatic tumors in the retroperitoneum, which were excised, digested, and cultured. Western blots, enzyme activity and enzyme activity gels were performed for manganese superoxide dismutase (MnSOD), copper/zinc (CuZnSOD), catalase, and glutathione peroxidase (GPx) in the ascites cell line, metastatic tumor cell line, MIA PaCa-2 primary pancreatic cancer cell line, and the Capan-1, a metastatic pancreatic cancer cell line. Cell growth, plating efficiency, growth in soft agar and growth in nude mice were determined in the ascites, metastatic tumor, and MIA PaCa-2 cell lines. MnSOD, CuZnSOD, and GPx protein and activity were increased in the ascites, metastatic tumor, and Capan-1 cell lines compared to MIA PaCa-2. The ascites and metastatic tumor cell lines had decreased cell growth, plating efficiency, and growth in soft agar, but the ascites cell line had increased cell growth in 4 and 1% O(2) concentrations in vitro and more rapid growth in vivo. Metastatic disease is associated with changes in the content and activity of antioxidant enzymes with an associated change in growth characteristics depending on the O(2) concentrations.
Collapse
Affiliation(s)
- Anne Lewis
- University of Iowa College of Medicine, Iowa City, 52242, USA
| | | | | | | | | | | |
Collapse
|
73
|
Martin RCG, Ahn J, Nowell SA, Hein DW, Doll MA, Martini BD, Ambrosone CB. Association between manganese superoxide dismutase promoter gene polymorphism and breast cancer survival. Breast Cancer Res 2006; 8:R45. [PMID: 16859522 PMCID: PMC1779484 DOI: 10.1186/bcr1532] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 06/13/2006] [Accepted: 07/11/2006] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Manganese superoxide dismutase (MnSOD) plays a critical role in the detoxification of mitochondrial reactive oxygen species, constituting a major cellular defense mechanism against agents that induce oxidative stress. A genetic polymorphism in the mitochondrial targeting sequence of this gene has been associated with increased cancer risk and survival in breast cancer. This base pair transition (-9 T > C) leads to a valine to alanine amino acid change in the mitochondrial targeting sequence. A polymorphism has also been identified in the proximal region of the promoter (-102 C>T) that alters the recognition sequence of the AP-2 transcription factor, leading to a reduction in transcriptional activity. The aim of our study was to investigate possible associations of the -102 C>T polymorphism with overall and relapse-free breast cancer survival in a hospital-based case-only study. MATERIALS AND METHODS The relationship between the MnSOD -102 C>T polymorphism and survival was examined in a cohort of 291 women who received chemotherapy and/or radiotherapy for incident breast cancer. The MnSOD -102 C>T genotype was determined using a TaqMan allele discrimination assay. Patient survival was evaluated according to the MnSOD genotype using Kaplan-Meier survival functions. Hazard ratios were calculated from adjusted Cox proportional hazards modeling. All statistical tests were two-sided. RESULTS In an evaluation of all women, there was a borderline significant reduction in recurrence-free survival with either one or both variant alleles (CT + TT) when compared with patients with wild-type alleles (CC) (odds ratio, 0.65; 95% confidence interval, 0.42-1.01). When the analysis was restricted to patients receiving radiation therapy, there was a significant reduction in relapse-free survival in women who were heterozygous for the MnSOD -102 genotype (relative risk, 0.40; 95% confidence interval, 0.18-0.86). Similarly, when the homozygous and heterozygous variant genotypes were combined, there remained a significant reduction in relapse-free survival in this group (hazard ratio, 0.42; 95% confidence interval, 0.20-0.87). CONCLUSION The MnSOD -102 variant allele appears to be associated with an improved recurrence-free survival in all patients, and more dramatically in subjects who received adjuvant radiation therapy.
Collapse
Affiliation(s)
- Robert CG Martin
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jiyoung Ahn
- Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA
- Department of Epidemiology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Susan A Nowell
- Division of Molecular Epidemiology, National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - David W Hein
- Department of Pharmacology and Toxicology, and the James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Mark A Doll
- Department of Pharmacology and Toxicology, and the James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Benjamin D Martini
- Department of Pharmacology and Toxicology, and the James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | |
Collapse
|
74
|
Malafa MP, Fokum FD, Andoh J, Neitzel LT, Bandyopadhyay S, Zhan R, Iiizumi M, Furuta E, Horvath E, Watabe K. Vitamin E succinate suppresses prostate tumor growth by inducing apoptosis. Int J Cancer 2006; 118:2441-7. [PMID: 16380976 DOI: 10.1002/ijc.21689] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Prostate cancer is a major cause of cancer death and morbidity in western countries. However, because of its intrinsic nature of chemoresistance, there is only limited systemic therapy available for the patients. Vitamin E (VE) has been under intensive study as a chemopreventive agent for various types of cancers. Preclinical studies suggest that vitamin E succinate (VES) is the most effective antitumor analogue of VE, yet there are scarce studies of VES in prostate cancer. In this study, we investigated the effects of VES on a panel of prostate cancer cells, and a xenograft model of prostate cancer. Our results indicate that VES significantly inhibited proliferation and induced apoptosis of prostate cancer cell lines in a dose and time dependent manner. The results of microarray analysis followed by real-time RT-PCR and inhibitor analyses indicated that the VES-induced apoptosis is mediated by caspase-4 in prostate tumor cells. In our animal model of prostate cancer in SCID mouse, daily injection of VES significantly suppressed tumor growth as well as lung metastases. These results suggest a potential therapeutic utility of VES for patients with prostate cancer.
Collapse
Affiliation(s)
- Mokenge P Malafa
- Division of GI Tumors, Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Behrend L, Mohr A, Dick T, Zwacka RM. Manganese superoxide dismutase induces p53-dependent senescence in colorectal cancer cells. Mol Cell Biol 2005; 25:7758-69. [PMID: 16107721 PMCID: PMC1190300 DOI: 10.1128/mcb.25.17.7758-7769.2005] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The mitochondrial enzyme manganese superoxide dismutase (MnSOD) is known to suppress cell growth in different tumor cell lines. However, the molecular mechanism of this growth-retarding effect is not fully understood. Here we show that overexpression of MnSOD slows down growth of HCT116 human colorectal cancer cells by induction of cellular senescence. MnSOD overexpression causes up-regulation of p53 and its transcriptional target, the cyclin-dependent kinase inhibitor p21. Adenovirus-mediated knockdown of p53 by RNA interference rescues MnSOD-overexpressing clones from growth retardation. Accordingly, the overexpression of MnSOD in HCTp53(-/-) cells does not lead to senescence, whereas in HCTp21(-/-) cells we found induction of senescence by forced expression of MnSOD. These results indicate a pivotal role of p53, but not p21, in the observed effects. Analysis of the mitochondrial membrane potential revealed reduced polarization in MnSOD-overexpressing cells. In addition, depolarization of the mitochondrial membrane by mitochondrial inhibitors such as rotenone or antimycin A led colorectal cancer cells into p53-dependent senescence. Our data indicate that uncoupling of the electrochemical gradient by increased MnSOD activity gives rise to p53 up-regulation and induction of senescence. This novel mitochondrially mediated mechanism of tumor suppression might enable strategies that allow reactivation of cellular aging in tumor cells.
Collapse
Affiliation(s)
- Lars Behrend
- Division of Gene Therapy, University of Ulm, Germany
| | | | | | | |
Collapse
|
76
|
Hu Y, Rosen DG, Zhou Y, Feng L, Yang G, Liu J, Huang P. Mitochondrial manganese-superoxide dismutase expression in ovarian cancer: role in cell proliferation and response to oxidative stress. J Biol Chem 2005; 280:39485-92. [PMID: 16179351 DOI: 10.1074/jbc.m503296200] [Citation(s) in RCA: 213] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Superoxide dismutases (SODs) are important antioxidant enzymes responsible for the elimination of superoxide radical (O(2)(-)). The manganese-containing SOD (Mn-SOD) has been suggested to have tumor suppressor function and is located in the mitochondria where the majority of O(2)(-) is generated during respiration. Although increased reactive oxygen species (ROS) in cancer cells has long been recognized, the expression of Mn-SOD in cancer and its role in cancer development remain elusive. The present study used a human tissue microarray to analyze Mn-SOD expression in primary ovarian cancer tissues, benign ovarian lesions, and normal ovary epithelium. Significantly higher levels of Mn-SOD protein expression were detected in the malignant tissues compared with normal tissues (p < 0.05). In experimental systems, suppression of Mn-SOD expression by small interfering RNA caused a 70% increase of superoxide in ovarian cancer cells, leading to stimulation of cell proliferation in vitro and more aggressive tumor growth in vivo. Furthermore, stimulation of mitochondrial O(2)(-) production induced an increase of Mn-SOD expression. Our findings suggest that the increase in Mn-SOD expression in ovarian cancer is a cellular response to intrinsic ROS stress and that scavenging of superoxide by SOD may alleviate the ROS stress and thus reduce the simulating effect of ROS on cell growth.
Collapse
Affiliation(s)
- Yumin Hu
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
77
|
Martin RCG, Lan Q, Hughes K, Doll MA, Martini BD, Lissowska J, Zatonski W, Rothman N, Hein DW. No apparent association between genetic polymorphisms (-102 C>T) and (-9 T>C) in the human manganese superoxide dismutase gene and gastric cancer(1). J Surg Res 2005; 124:92-7. [PMID: 15734485 DOI: 10.1016/j.jss.2004.09.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2004] [Indexed: 01/03/2023]
Abstract
BACKGROUND Manganese superoxide dismutase (MnSOD) plays a critical role in the detoxification of mitochondrial reactive oxygen species, constituting a major cellular defense mechanism against agents that induce oxidative stress. A genetic polymorphism in the mitochondrial targeting sequence of this gene has been associated with increased cancer risk. This one base pair transition (-9 T>C) leads to a Val to Ala amino acid change in the mitochondrial targeting sequence. In addition, the MnSOD promoter contains an activator protein-2 (AP-2) binding site that modifies transcription of MnSOD. Mutations have been identified in the proximal region of the promoter in human tumor cell lines. One of these mutations (-102 C>T) has been shown to change the binding pattern of AP-2, leading to a reduction in transcriptional activity. The aim of our study was to investigate possible associations of the (-9 T>C) and (-102 C>T) polymorphisms with gastric cancer in a population-based case-control study conducted in Warsaw, Poland. MATERIALS AND METHODS DNA was obtained from a population based case-control study of stomach cancer conducted in Warsaw, Poland, between 1994 and 1996. The MnSOD -9 T>C genotype was determined by PCR-RFLP assay. The MnSOD -102 C>T genotype was determined using a TaqMan allele discrimination assay. RESULTS The frequency of the -102 C>T polymorphism was 41% (38/91) in gastric cancer cases and 38% (50/130) in the controls (odds ratio [OR] 1.1, 95% confidence interval [CI] 0.6-2.1). The frequency of the -9 T>C polymorphism was 44% (202/464) in cases and 56% (262/464) in controls (OR 1.1; 95% CI 0.9-1.37). The lack of association was observed in both non-smokers (OR 1.5; 95% CI 0.7-2.34) and smokers (OR 1.1; 95% CI 0.7-1.7). Furthermore, the association was not significant when smokers were segregated by extent of smoking history. CONCLUSION The association of the manganese superoxide dismutase polymorphisms at -102 C>T and the -9 T>C were not found to be associated with gastric cancer in a Polish case-control study.
Collapse
Affiliation(s)
- Robert C G Martin
- Department of Surgery, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Abstract
The role of antioxidant enzymes can be interpreted in terms of fine tuning of the concentration of reactive oxygen species which are required in the redox regulation of the cell cycle and of programmed cell death. This review summarizes findings from papers published in the last few years which deal with the relation between apoptosis and the two antioxidant enzymes, manganous superoxide dismutase (MnSOD) and catalase. With respect to MnSOD, the literature is much in favor of an inhibitory action in apoptosis. Increased MnSOD activity has been shown to prevent cell death via the receptor-mediated apoptotic pathway as well as cell death via the mitochondrial pathway. The literature on the influence of catalase activity on apoptosis is less consistent. Evidence for both an antiapoptotic and a proapoptotic role of catalase can be found. From the results reviewed here, two schemes for the involvement of MnSOD and catalase in the regulation of apoptosis can be extracted: 1) Both MnSOD and catalase inhibit apoptosis by removing superoxide anion radicals or H2O2, respectively, because these reactive oxygen species are mediators required for the apoptotic program or inhibit a survival pathway. 2) An increase in H2O2 by downregulation or inhibition of catalase activity and/or upregulation of MnSOD activity inhibits apoptosis while a decrease in H2O2 by upregulation of catalase activity and/or downregulation of MnSOD activity supports apoptosis, possibly because of a supportive role of H2O2 in a survival pathway. The data reported so far do not allow for an explanation why some cell models appear to fit the first scheme while the second scheme appears to correctly describe other cell models. The present state of the literature reveals that antioxidant enzymes play a more intricate role in cell physiology than previously assumed.
Collapse
Affiliation(s)
- Regine Kahl
- Institute of Toxicology, Heinrich Heine University, Düsseldorf, Germany.
| | | | | | | |
Collapse
|
79
|
Li H, Kantoff PW, Giovannucci E, Leitzmann MF, Gaziano JM, Stampfer MJ, Ma J. Manganese Superoxide Dismutase Polymorphism, Prediagnostic Antioxidant Status, and Risk of Clinical Significant Prostate Cancer. Cancer Res 2005; 65:2498-504. [PMID: 15781667 DOI: 10.1158/0008-5472.can-04-3535] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oxidative stress may enhance prostatic carcinogenesis. A polymorphism [valine (V) --> alanine (A)] of manganese superoxide dismutase (MnSOD), the primary antioxidant enzyme in mitochondria, has been recently associated with prostate cancer. We examined the relationship between prostate cancer and the MnSOD polymorphism and its interactions with baseline plasma antioxidant levels (selenium, lycopene, and alpha-tocopherol) and beta-carotene treatment among 567 cases and 764 controls nested in the prospective Physicians' Health Study. We found little overall association between MnSOD polymorphism and prostate cancer risk; however, this polymorphism significantly modified risk of prostate cancer associated with prediagnostic plasma antioxidants (P(interaction) > or = 0.05). Among men with the AA genotype, high selenium level (4th versus 1st quartile) was associated with a relative risk (RR) of 0.3 [95% confidence interval (CI), 0.2-0.7] for total prostate cancer; for clinically aggressive prostate cancer, the RR was 0.2 (95% CI, 0.1-0.5). In contrast, among men with the VV/VA genotype, the RRs were 0.6 (0.4-1.0) and 0.7 (0.4-1.2) for total and clinically aggressive prostate cancer. These patterns were similar for lycopene and alpha-tocopherol and were particularly strong when these antioxidants and selenium were combined; men with the AA genotype had a 10-fold gradient in risk for aggressive prostate cancer across quartiles of antioxidant status. Men with AA genotype who were randomly assigned to beta-carotene treatment (versus placebo) had a RR of 0.6 (95% CI, 0.2-0.9; P(interaction) = 0.03) for fatal prostate cancer, but no significant association was observed in men with the VV/VA genotype. Both endogenous and exogenous antioxidants play an important and interdependent role in preventing clinically significant prostate cancer.
Collapse
Affiliation(s)
- Haojie Li
- Channing Laboratory, Department of Medicine Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
80
|
Nishikawa M, Tamada A, Hyoudou K, Umeyama Y, Takahashi Y, Kobayashi Y, Kumai H, Ishida E, Staud F, Yabe Y, Takakura Y, Yamashita F, Hashida M. Inhibition of experimental hepatic metastasis by targeted delivery of catalase in mice. Clin Exp Metastasis 2004; 21:213-21. [PMID: 15387371 DOI: 10.1023/b:clin.0000037706.13747.5e] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Bovine liver catalase derivatives possessing diverse tissue distribution properties were synthesized, and their effects on hepatic metastasis of colon carcinoma cells were examined in mice. An intraportal injection of 1 x 10(5) colon 26 cells resulted in the formation of more than 50 metastatic colonies on the surface of the liver at 14 days after injection. An intravenous injection of catalase (CAT; 35000 units/kg of body weight) significantly (P < 0.001) reduced the number of the colonies in the liver. Galactosylated (Gal-), mannosylated (Man-) and succinylated (Suc-) CAT were also tested in the same system. Of these derivatives, Gal-CAT showed the greatest inhibitory effect on hepatic metastasis, and the number of colonies was significantly (P < 0.001) smaller than following treatment with catalase. High activities of matrix metalloproteinases (MMPs), especially MMP-9, were detected in the liver of mice bearing metastatic tumor tissues, which was significantly (P < 0.05) reduced by Gal-CAT. These results, combined with our previous finding that Gal-CAT can be efficiently delivered to hepatocytes, indicate that the targeted delivery of catalase to the liver by galactosylation is a promising approach to suppress hepatic metastasis. Decreased MMP activity by catalase delivery seems to be involved in its anti-metastatic effect.
Collapse
Affiliation(s)
- Makiya Nishikawa
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Nelson KK, Melendez JA. Mitochondrial redox control of matrix metalloproteinases. Free Radic Biol Med 2004; 37:768-84. [PMID: 15304253 DOI: 10.1016/j.freeradbiomed.2004.06.008] [Citation(s) in RCA: 336] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Revised: 05/17/2004] [Accepted: 06/04/2004] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are constantly generated in aerobic organisms during normal metabolism and in response to both internal and external stimuli. Imbalances in the production and removal of ROS have been hypothesized to play a causative role in numerous disease pathologies such as cancer, ischemia/reperfusion injury, and degenerative diseases such as photoaging, atherosclerosis, arthritis, and neurodegeneration. A feature often associated with these diseases is a malfunctioning of the connective tissue remodeling process due to increased activity of extracellular matrix-degrading metalloproteinases (MMPs). This review summarizes the evidence that implicates ROS as key regulators of MMP production and the importance of these interactions in disease pathologies.
Collapse
Affiliation(s)
- Kristin K Nelson
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | | |
Collapse
|
82
|
Kinnula VL, Crapo JD. Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med 2004; 36:718-44. [PMID: 14990352 DOI: 10.1016/j.freeradbiomed.2003.12.010] [Citation(s) in RCA: 243] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Revised: 12/01/2003] [Accepted: 12/12/2003] [Indexed: 02/06/2023]
Abstract
Reactive oxygen metabolites have multifactorial effects on the regulation of cell growth and the capacity of malignant cells to invade. Overexpression of the superoxide dismutases (SODs) in vitro increases cell differentiation, decreases cell growth and proliferation, and can reverse a malignant phenotype to a nonmalignant one. The situation in vivo is more complex due to multiple interactions of tumor cells with their environment. Numerous in vivo studies show that the superoxide dismutases can be highly expressed in aggressive human solid tumors. Furthermore, high SOD has occasionally been associated with a poor prognosis and with resistance to cytotoxic drugs and radiation. Most of the apparent conflicts between the above in vitro and in vivo observations can be reconciled by considering the net redox status of tumor cells in different environments. Administering high concentrations of SOD to cells in vitro is usually associated with a non- or less malignant phenotype, whereas secondary induction of SOD in tumors in vivo can be associated with an aggressive malignant transformation probably due to the altered (oxidative) redox state in the malignant cells. This concept suggests that for many types of tumors antioxidants could be used to diminish the invasive capability of malignant cells.
Collapse
Affiliation(s)
- Vuokko L Kinnula
- Division of Pulmonary Medicine, Department of Medicine, University of Helsinki and Helsinki University Hospital, FIN-0029 Helsinki, Finland.
| | | |
Collapse
|
83
|
Mukhopadhyay S, Das SK, Mukherjee S. Expression of Mn-Superoxide Dismutase Gene in Nontumorigenic and Tumorigenic Human Mammary Epithelial Cells. J Biomed Biotechnol 2004; 2004:195-202. [PMID: 15467159 PMCID: PMC555772 DOI: 10.1155/s1110724304401016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Manganese superoxide dismutase (Mn-SOD), localized
at the mitochondrial matrix, has the ability to protect cells
against oxidative damage. It has been reported that low levels of
Mn-SOD gene expression cause the development of certain kind of
tumors. On the other hand, overexpression of Mn-SOD gene may play
an important role in the development of cancer. In our study, we
find that Mn-SOD activity was higher in nonaggressive (MCF-7) and
aggressive (BT-549 and 11-9-14) breast cancer cell lines compared
to that of nontumorigenic (MCF-12A and MCF-12F) mammary
epithelial cell lines. We also observed an increased expression
of Mn-SOD gene in cancerous cell lines. The elevated level of SOD
activity in nonaggressive and aggressive breast epithelial cell
lines was associated with some changes in nucleotide sequence.
Collapse
Affiliation(s)
- Sutapa Mukhopadhyay
- Department of Pharmacology, Meharry Medical College,
Nashville, TN 37208, USA
| | - Salil K. Das
- Department of Biochemistry, Meharry Medical College,
Nashville, TN 37208, USA
- *Salil K. Das:
| | - Shyamali Mukherjee
- Department of Pharmacology, Meharry Medical College,
Nashville, TN 37208, USA
| |
Collapse
|
84
|
Park SY, Chang I, Kim JY, Kang SW, Park SH, Singh K, Lee MS. Resistance of mitochondrial DNA-depleted cells against cell death: role of mitochondrial superoxide dismutase. J Biol Chem 2003; 279:7512-20. [PMID: 14660625 DOI: 10.1074/jbc.m307677200] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We have shown that mitochondrial DNA-depleted (rho(0)) SK-Hep1 hepatoma cells are resistant to apoptosis, contrary to previous papers reporting normal apoptotic susceptibility of rho(0) cells. We studied the changes of gene expression in SK-Hep1 rho(0) cells. DNA chip analysis showed that MnSOD expression was profoundly increased in rho(0) cells. O(2)(.) contents increased during rho(0) cell derivation but became normalized after establishment of rho(0) phenotypes, suggesting that MnSOD induction is an adaptive process to increased O(2)(.). rho(0) cells were resistant to menadione, paraquat, or doxorubicin, and O(2)(.) contents after treatment with them were lower in rho(0) cells compared with parental cells because of MnSOD overexpression. Expression levels and activity of glutathione peroxidases were also increased in rho(0) cells, rendering them resistant to exogenous H(2)O(2). rho(0) cells were resistant to p53, and intracellular ROS contents after p53 expression were lower compared with parental cells. Other types of rho(0) cells also showed increased MnSOD expression and resistance against ROS. Heme oxygenase-1 expression was increased in rho(0) cells, and a heme oxygenase-1 inhibitor decreased the induction of MnSOD in rho(0) cells and their resistance against ROS donors. These results indicate that rho(0) cells are resistant to cell death contrary to previous reports and suggest that an adaptive increase in the expression of antioxidant enzymes renders cancer cells or aged cells with frequent mitochondrial DNA mutations to resist against oxidative stress, host anti-cancer surveillance, or chemotherapeutic agents, conferring survival advantage on them.
Collapse
Affiliation(s)
- Sun Young Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwondong Kangnam-ku, Seoul 135-710, Korea
| | | | | | | | | | | | | |
Collapse
|
85
|
Schoenfeld N, Bauer MKA, Grimm S. The metastasis suppressor gene C33/CD82/KAI1 induces apoptosis through reactive oxygen intermediates. FASEB J 2003; 18:158-60. [PMID: 14597553 DOI: 10.1096/fj.03-0420fje] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Here we describe the isolation of C33/CD82/KAI1 in a screen for apoptosis-inducing genes. C33 is a gene that is downregulated in many metastatic tumor cells and the expression of which can attenuate the process of metastases formation in a variety of tumors. In accordance, we observed cell death induction by C33 in many different cell types. C33 seems to promote cell death by the generation of reactive oxygen intermediates (ROIs). These ROIs, however, are not derived from the mitochondrial respiratory chain as in most other scenarios leading to apoptosis. We observed that C33 renders cells sensitive to ROIs by causing the specific release of the intracellular antioxidant glutathione (GSH) from cells. Moreover, C33 activates the GTPase Cdc42, which mediates GSH release and apoptosis induction and allows to detect the formation of ROIs.
Collapse
|
86
|
Kyriazanos ID, Dhar DK, Rahman MA, Ohno S, Kubota H, Kohno H, Nagasue N. Impact of uncoupling protein-2 expression on the prognosis of gastric adenocarcinoma patients. Dig Liver Dis 2003; 35:597-9. [PMID: 14567467 DOI: 10.1016/s1590-8658(03)00275-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
87
|
Lin Y, Kikuchi S, Obata Y, Yagyu K. Serum copper/zinc superoxide dismutase (Cu/Zn SOD) and gastric cancer risk: a case-control study. Jpn J Cancer Res 2002; 93:1071-5. [PMID: 12417035 PMCID: PMC5926885 DOI: 10.1111/j.1349-7006.2002.tb01207.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We conducted a case-control study to evaluate the association between serum levels of copper/zinc superoxide dismutase (Cu/Zn SOD) and the risk of gastric cancer. Cases were 214 patients who had been diagnosed with gastric cancer and controls were 120 persons who underwent medical checkups. Serum levels of Cu/Zn SOD were determined by enzyme-linked immunosorbent assay (ELISA). Compared with the lowest quartile, the OR (odds ratio) was 4.54 (95% CI (confidence interval), 1.62 - 12.66) for the third quartile and 15.75 (95% CI, 5.84 - 42.46) for the highest quartile. With both early and advanced cancers, as well as with the intestinal and diffuse types, a significant increase in risk was observed with increasing levels of serum Cu/Zn SOD. Our case-control study showed that serum levels of Cu/Zn SOD were significantly elevated in gastric cancer patients compared with apparently healthy controls, and higher Cu/Zn SOD levels may be associated with an increased risk of gastric cancer.
Collapse
Affiliation(s)
- Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, 21 Karimata, Yazako, Nagakute-cho, Aichi-gun, Aichi 480-1195, Japan.
| | | | | | | |
Collapse
|
88
|
Piyathilake CJ, Bell WC, Oelschlager DK, Heimburger DC, Grizzle WE. The pattern of expression of Mn and Cu-Zn superoxide dismutase varies among squamous cell cancers of the lung, larynx, and oral cavity. Head Neck 2002; 24:859-67. [PMID: 12211050 DOI: 10.1002/hed.10135] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Despite the importance of reactive oxygen species (ROS) in the development of smoking-related cancers, little is known about the pattern of expression of ROS scavengers in these cancers. METHODS In this present study, we examined the expression of manganese superoxide dismutase (Mn-SOD) and copper/zinc superoxide dismutase (Cu-Zn-SOD), which are essential enzymes that eliminate ROS, in squamous cell cancers (SCCs) of the lung (n = 12), larynx (n = 13), and oral cavity (n = 20). RESULTS SCCs of larynx and oral cavity showed significantly enhanced immuhistochemical expression of Mn-SOD compared with the matched uninvolved epithelium. The higher expression of Mn-SOD was shown to be late and early events in the process of SCC development in the larynx and the oral cavity, respectively. The expression of Mn-SOD in SCCs of the lung was significantly lower compared with luminal cells of the uninvolved epithelium but not compared with basal cells or an average expression of SOD in basal and luminal cells. The expression of both Mn-SOD and cytoplasmic or nuclear Cu-Zn-SOD in bronchial epithelium adjacent to invasive cancer was significantly lower compared with its expression in the uninvolved bronchial epithelium away from cancer. This resulted in a significant difference in SOD expression between cancer and uninvolved bronchial epithelium away from cancer but not between cancer and uninvolved epithelium adjacent to cancer. CONCLUSIONS There are significant differences in the expression of Mn-SOD and Cu-Zn-SOD among SCCs of the lung, larynx, and oral cavity. The results also suggest that variations in distance between cancer and uninvolved tissues evaluated could contribute to conflicting results of SOD expression.
Collapse
Affiliation(s)
- Chandrika J Piyathilake
- Division of Nutritional Biochemistry and Molecular Biology, University of Alabama at Birmingham, University Station, Birmingham, Alabama 35294, USA.
| | | | | | | | | |
Collapse
|
89
|
Abstract
BACKGROUND Vitamin E succinate (VES) is a promising anti-cancer micronutrient. In this study, we tested the hypothesis that VES will promote colon cancer tumor dormancy and inhibit liver metastases in colon cancer. METHODS CT-26 colon cancer cells were treated with VES in vitro and in an in vivo model of liver metastases. The impact of VES on cellular proliferation and apoptosis was measured in vitro by MTS assay and sandwich ELISA and in vivo by PCNA staining and TUNEL assay, respectively. Correlation coefficients and independent t tests were used for statistical analysis. RESULTS VES significantly and specifically inhibited cell proliferation (P = 0.011) and promoted apoptosis (P < 0.0074) of cancer cells in vitro. VES produced a 40% reduction of liver metastases (P = 0.037). Five of the eight mice had an excellent response to VES. Subsequent analysis of these five mice revealed a 75% reduction in the number of liver metastases (P < 0.05). VES significantly promoted tumor cell apoptosis (P < 0.0003) and inhibited cell proliferation (P = 0.0069) in vivo. CONCLUSIONS VES inhibits the growth of colon cancer cells in vitro and in vivo. This is the first report of VES inhibition of colon cancer tumor metastases. The mechanism of VES anti-tumor and anti-metastatic activity in vivo appears to involve promotion of tumor apoptosis and inhibition of cell proliferation. These findings support further investigation of VES as a micronutrient to promote colon cancer tumor dormancy and prevent metastases.
Collapse
Affiliation(s)
- Kevin T Barnett
- Department of Surgery, Southern Illinois University School of Medicine, Springfield 62794, USA
| | | | | |
Collapse
|
90
|
Zhang HJ, Zhao W, Venkataraman S, Robbins MEC, Buettner GR, Kregel KC, Oberley LW. Activation of matrix metalloproteinase-2 by overexpression of manganese superoxide dismutase in human breast cancer MCF-7 cells involves reactive oxygen species. J Biol Chem 2002; 277:20919-26. [PMID: 11929863 DOI: 10.1074/jbc.m109801200] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) participate in cell migration and remodeling processes by affecting the extracellular matrix. MMP-2 is thought to be involved in cancer cell invasiveness. It has been proposed that the activity of MMP-2 can be modulated by intracellular reactive oxygen species (ROS)/reactive nitrogen species. We hypothesized that manganese superoxide dismutase (MnSOD) could mediate MMP-2 activity by changing the intracellular ROS level and that nitric oxide ((.)NO) may be involved in this process. Human breast cancer MCF-7 cells were stably transfected with plasmids containing MnSOD cDNA. A 2-30-fold increase of MnSOD protein and activity was observed in four clones. Our data demonstrated that overexpression of MnSOD stimulated the activation of MMP-2 with a corresponding elevation of ROS. A decrease in ROS by ebselen, a glutathione peroxidase mimetic, or by transduction of adenovirus containing human catalase or glutathione peroxidase cDNA abolished the effect of MnSOD on MMP-2 activation. Treatment of MCF-7 cells with antimycin A or rotenone increased intracellular ROS production and MMP-2 activation simultaneously. Our data also showed a suppression of endothelial nitric-oxide synthase expression that was accompanied by decreased (.)NO production in MnSOD-overexpressing cells. However, the changes in endothelial nitric-oxide synthase and (.)NO did not correlate with the MnSOD activity. Corresponding changes of MMP-2 activity after the addition of a NOS inhibitor (N(G)-amino-l-arginine) or a (.)NO donor ((Z)-1-[(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate) to the cells suggested the possibility that (.)NO may be involved in the MnSOD-mediated MMP-2 activation pathway. These results indicate that MnSOD induces MMP-2 activity by regulation of intracellular ROS and imply that signaling pathways involving (.)NO may also be involved in the MnSOD mediation of MMP-2 activity.
Collapse
Affiliation(s)
- Hannah J Zhang
- Department of Exercise Science and Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
91
|
Abstract
BACKGROUND Previous work has demonstrated that vitamin E succinate (VES), an ester analogue of vitamin E, inhibits the growth of melanoma in vitro. However, there is no information about the effect of VES on melanoma in vivo. We investigated the effect of VES on melanoma in vitro and in vivo. METHODS The effect of VES on the proliferation and apoptosis of the B16F10 murine melanoma cell line was determined by a modified Cell Titer 96 AQ assay and a cell death detection enzyme-linked immunosorbent assay, respectively. The in vivo effect of VES on B16F10 melanoma cells allografted in athymic nude mice was investigated. The mechanism of the in vivo antitumor effect of VES was determined by immunohistochemical detection of proliferation and apoptosis. RESULTS VES decreased cell proliferation (P =.0001) and increased cell apoptosis (P =.0001) in a dose-dependent manner in vitro. Also, VES significantly inhibited melanoma growth in mice (P =.0013). The VES antitumor effect in vivo was associated with a significant increase in the melanoma apoptosis rate (P =.0256). CONCLUSIONS This is the first report of the antimelanoma effect of VES in vivo. The mechanism of the antimelanoma effect of VES in vivo involves the promotion of tumor cell apoptosis. These findings support future investigations of VES as a therapeutic micronutrient against melanoma.
Collapse
Affiliation(s)
- Mokenge P Malafa
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, IL 62794-9638, USA
| | | | | | | | | |
Collapse
|
92
|
Ranganathan AC, Nelson KK, Rodriguez AM, Kim KH, Tower GB, Rutter JL, Brinckerhoff CE, Huang TT, Epstein CJ, Jeffrey JJ, Melendez JA. Manganese superoxide dismutase signals matrix metalloproteinase expression via H2O2-dependent ERK1/2 activation. J Biol Chem 2001; 276:14264-70. [PMID: 11297530 DOI: 10.1074/jbc.m100199200] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Manganese-superoxide dismutase (Sod2) removes mitochondrially derived superoxide (O(2)) at near-diffusion limiting rates and is the only antioxidant enzyme whose expression is regulated by numerous stimuli. Here it is shown that Sod2 also serves as a source of the intracellular signaling molecule H(2)O(2). Sod2-dependent increases in the steady-state levels of H(2)O(2) led to ERK1/2 activation and subsequent downstream transcriptional increases in matrix metalloproteinase-1 (MMP-1) expression, which were reversed by expression of the H(2)O(2)-detoxifying enzyme, catalase. In addition, a single nucleotide polymorphism has recently been identified (1G/2G) at base pair--1607 that creates an Ets site adjacent to an AP-1 site at base pair --1602 and has been shown to dramatically enhance transcription of the MMP-1 promoter. Luciferase promoter constructs containing either the 1G or 2G variation were 25- or 1000-fold more active when transiently transfected into Sod2-overexpressing cell lines, respectively. The levels of MMP-2, -3, and -7 were also increased in the Sod2-overexpressing cell lines, suggesting that Sod2 may function as a "global" redox regulator of MMP expression. In addition, Sod2(-/+) mouse embryonic fibroblasts failed to respond to the cytokine-mediated induction of the murine functional analog of MMP-1, MMP-13. This study provides evidence that the modulation of Sod2 activity by a wide array of pathogenic and inflammatory stimuli may be utilized by the cell as a primary signaling mechanism leading to matrix metalloproteinase expression.
Collapse
Affiliation(s)
- A C Ranganathan
- Centers for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Tanaka M, Kogawa K, Nakamura K, Nishihori Y, Kuribayashi K, Hagiwara S, Muramatsu H, Sakamaki S, Niitsu Y. Anti-metastatic gene therapy utilizing subcutaneous inoculation of EC-SOD gene transduced autologous fibroblast suppressed lung metastasis of Meth-A cells and 3LL cells in mice. Gene Ther 2001; 8:149-56. [PMID: 11313784 DOI: 10.1038/sj.gt.3301362] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2000] [Accepted: 09/14/2000] [Indexed: 11/09/2022]
Abstract
We have previously reported that superoxide stimulates the motility of tumor cells and the administration of Cu-Zn superoxide dismutase (SOD) significantly suppresses metastasis. However, ideally, anti-metastatic therapy should be long-lasting, systemically effective and have low toxicity. The half-life of Cu-Zn SOD in plasma is so short that it cannot provide long-lasting effects. Therefore, in this study we have developed a gene therapy in a mouse model utilizing extracellular SOD (EC-SOD), which is the most prevalent SOD isoenzyme in extracellular fluids. We retrovirally transfected fibroblasts (syngeneic) with the EC-SOD gene and established EC-SOD-secreting fibroblasts. Inoculation of EC-SOD-secreting fibroblasts suppressed both artificial and spontaneous metastatic lung nodules in mouse metastasis models. These data indicate the feasibility of anti-metastatic gene therapy utilizing the EC-SOD gene.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/secondary
- Carcinoma, Lewis Lung/therapy
- Cell Culture Techniques
- Cell Division
- Culture Media
- DNA, Complementary/genetics
- Feasibility Studies
- Fibroblasts/transplantation
- Gene Expression
- Genetic Therapy/methods
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred Strains
- Neoplasm Transplantation
- RNA, Messenger/genetics
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/secondary
- Sarcoma, Experimental/therapy
- Superoxide Dismutase/genetics
- Superoxide Dismutase/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- M Tanaka
- Fourth Department of Internal Medicine, Sapporo Medical University School of Medicine, South-1, West-16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
|
95
|
Abstract
BACKGROUND Vitamin E succinate (VES) is the most potent antitumor analogue of vitamin E. Despite many reports of VES's antitumor activity in vitro, there is little information about its antitumor effects in vivo. MATERIALS AND METHODS We investigated the effect of VES on the growth of human breast cancer cells in vitro and in vivo. RESULTS VES decreased cell viability in MDA-MB-231 and MCF-7 human breast cancer cells. Although VES increased apoptosis in MDA-MB-231 cells, it had no effect on apoptosis in MCF-7 cells. The inhibitory effect of VES on cell growth was specific for the intact molecule because a markedly reduced effect was noted when either vitamin E or succinic acid was administered alone. VES inhibited the growth of MDA-MB-231 cells in nude mice. Also, VES was found to inhibit vascular endothelial growth factor (VEGF) gene expression in MDA-MB-231 cells. CONCLUSIONS VES inhibits the growth of breast cancer cells in vitro and in vivo. This is the first report of VES inhibition of established tumor growth in vivo. The mechanism of VES's in vivo effects may involve inhibition of tumor angiogenesis since VES inhibits VEGF gene expression.
Collapse
Affiliation(s)
- M P Malafa
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois 62794, USA.
| | | |
Collapse
|