251
|
Idh1 protects murine hepatocytes from endotoxin-induced oxidative stress by regulating the intracellular NADP(+)/NADPH ratio. Cell Death Differ 2015; 22:1837-45. [PMID: 25882048 PMCID: PMC4648331 DOI: 10.1038/cdd.2015.38] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 01/11/2023] Open
Abstract
Isocitrate dehydrogenase-1 (Idh1) is an important metabolic enzyme that produces NADPH by converting isocitrate to α-ketoglutarate. Idh1 is known to reduce reactive oxygen species (ROS) induced in cells by treatment with lipopolysaccharide (LPS) in vitro. Here, we used Idh1-deficient knockout (Idh1 KO) mice to investigate the role of Idh1 in antioxidant defense in vivo. Idh1 KO mice showed heightened susceptibility to death induced by LPS and exhibited increased serum levels of inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. The serum of LPS-injected Idh1 KO mice also contained elevated levels of AST, a marker of inflammatory liver damage. Furthermore, after LPS injection, livers of Idh1 KO mice showed histological evidence of elevated oxidative DNA damage compared with livers of wild-type (WT) mice. Idh1 KO livers showed a faster and more pronounced oxidative stress than WT livers. In line with that, Idh1 KO hepatocytes showed higher ROS levels and an increase in the NADP(+)/NADPH ratio when compared with hepatocytes isolated from WT mice. These results suggest that Idh1 has a physiological function in protecting cells from oxidative stress by regulating the intracellular NADP(+)/NADPH ratio. Our findings suggest that stimulation of Idh1 activity may be an effective therapeutic strategy for reducing oxidative stress during inflammatory responses, including the early stages of septic shock.
Collapse
|
252
|
Oxidative stress responses and NRF2 in human leukaemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:454659. [PMID: 25918581 PMCID: PMC4396545 DOI: 10.1155/2015/454659] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/15/2015] [Accepted: 03/20/2015] [Indexed: 01/21/2023]
Abstract
Oxidative stress as a result of elevated levels of reactive oxygen species (ROS) has been observed in almost all cancers, including leukaemia, where they contribute to disease development and progression. However, cancer cells also express increased levels of antioxidant proteins which detoxify ROS. This includes glutathione, the major antioxidant in human cells, which has recently been identified to have dysregulated metabolism in human leukaemia. This suggests that critical balance of intracellular ROS levels is required for cancer cell function, growth, and survival. Nuclear factor (erythroid-derived 2)-like 2 (NRF2) transcription factor plays a dual role in cancer. Primarily, NRF2 is a transcription factor functioning to protect nonmalignant cells from malignant transformation and oxidative stress through transcriptional activation of detoxifying and antioxidant enzymes. However, once malignant transformation has occurred within a cell, NRF2 functions to protect the tumour from oxidative stress and chemotherapy-induced cytotoxicity. Moreover, inhibition of the NRF2 oxidative stress pathway in leukaemia cells renders them more sensitive to cytotoxic chemotherapy. Our improved understanding of NRF2 biology in human leukaemia may permit mechanisms by which we could potentially improve future cancer therapies. This review highlights the mechanisms by which leukaemic cells exploit the NRF2/ROS response to promote their growth and survival.
Collapse
|
253
|
Nohara K, Okamura K, Suzuki T, Murai H, Ito T, Shinjo K, Takumi S, Michikawa T, Kondo Y, Hata K. Augmenting effects of gestational arsenite exposure of C3H mice on the hepatic tumors of the F₂ male offspring via the F₁ male offspring. J Appl Toxicol 2015; 36:105-12. [PMID: 25825268 DOI: 10.1002/jat.3149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/13/2015] [Accepted: 02/17/2015] [Indexed: 02/06/2023]
Abstract
Gestational exposure can affect the F2 generation through exposure of F1 germline cells. Previous studies reported that arsenite exposure of only F0 females during their pregnancy increases hepatic tumors in the F1 males in C3H mice, whose males are predisposed spontaneously to develop hepatic tumors later in life. The present study addressed the effects of gestational arsenite exposure on tumorigenesis of the F2 males in C3H mice. Expression analysis of several genes in the normal livers at 53 and 80 weeks of age clearly showed significant changes in the F2 males obtained by crossing gestational arsenite-exposed F1 (arsenite-F1) males and females compared to the control F2 males. Some of the changes were shown to occur in a late-onset manner. Then the tumor incidence was assessed at 75-82 weeks of age in the F2 males obtained by reciprocal crossing between the control and arsenite-F1 males and females. The results demonstrated that the F2 males born to arsenite-F1 males developed tumors at a significantly higher rate than the F2 males born to the control F1 males, irrespective of exposure of F1 females. Gene expressions of hepatocellular carcinoma markers β-catenin (CTNNB1) and interleukin-1 receptor antagonist in the tumors were significantly upregulated in the F2 males born to arsenite-F1 males compared to those born to the control F1 males. These results show that arsenite exposure of only F0 pregnant mice causes late-onset changes and augments tumors in the livers of the F2 males by affecting the F1 male offspring.
Collapse
Affiliation(s)
- Keiko Nohara
- Center for Environmental Health Sciences, National Institute for Environmental Studies, Tsukuba, Japan
| | - Kazuyuki Okamura
- Center for Environmental Health Sciences, National Institute for Environmental Studies, Tsukuba, Japan
| | - Takehiro Suzuki
- Center for Environmental Health Sciences, National Institute for Environmental Studies, Tsukuba, Japan
| | - Hikari Murai
- Center for Environmental Health Sciences, National Institute for Environmental Studies, Tsukuba, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Keiko Shinjo
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shota Takumi
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takehiro Michikawa
- Center for Environmental Health Sciences, National Institute for Environmental Studies, Tsukuba, Japan
| | - Yutaka Kondo
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
254
|
Hung N, Shen CC, Hu YW, Hu LY, Yeh CM, Teng CJ, Kuan AS, Chen SC, Chen TJ, Liu CJ. Risk of cancer in patients with iron deficiency anemia: a nationwide population-based study. PLoS One 2015; 10:e0119647. [PMID: 25781632 PMCID: PMC4363660 DOI: 10.1371/journal.pone.0119647] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
Objective This study evaluated the risk of cancer among patients with iron deficiency anemia (IDA) by using a nationwide population-based data set. Method Patients newly diagnosed with IDA and without antecedent cancer between 2000 and 2010 were recruited from the Taiwan National Health Insurance Research Database. The standardized incidence ratios (SIRs) of cancer types among patients with IDA were calculated. Results Patients with IDA exhibited an increased overall cancer risk (SIR: 2.15). Subgroup analysis showed that patients of both sexes and in all age groups had an increased SIR. After we excluded patients diagnosed with cancer within the first and first 5 years of IDA diagnosis, the SIRs remained significantly elevated at 1.43 and 1.30, respectively. In addition, the risks of pancreatic (SIR: 2.31), kidney (SIR: 2.23), liver (SIR: 1.94), and bladder cancers (SIR: 1.74) remained significantly increased after exclusion of patients diagnosed with cancer within 5 years after IDA diagnosis. Conclusion The overall cancer risk was significantly elevated among patients with IDA. After we excluded patients diagnosed with IDA and cancer within 1 and 5 years, the SIRs remained significantly elevated compared with those of the general population. The increased risk of cancer was not confined to gastrointestinal cancer when the SIRs of pancreatic, kidney, liver, and bladder cancers significantly increased after exclusion of patients diagnosed with IDA and cancer within the first 5 years. This finding may be caused by immune activities altered by IDA. Further study is necessary to determine the association between IDA and cancer risk.
Collapse
Affiliation(s)
- Ning Hung
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Che Shen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Psychiatry, Chiayi Branch, Taichung Veterans General Hospital, Chiayi, Taiwan
| | - Yu-Wen Hu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of information magagement, National Chung-Cheng University, Chiayi, Taiwan
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Yu Hu
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Chiu-Mei Yeh
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Jen Teng
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Oncology and Hematology, Department of Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Ai-Seon Kuan
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - San-Chi Chen
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Jen Liu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
255
|
Kumar R, Deep G, Agarwal R. An Overview of Ultraviolet B Radiation-Induced Skin Cancer Chemoprevention by Silibinin. ACTA ACUST UNITED AC 2015; 1:206-215. [PMID: 26097804 DOI: 10.1007/s40495-015-0027-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skin cancer incidences are rising worldwide, and one of the major causative factors is excessive exposure to solar ultraviolet radiation (UVR). Annually, ~5 million skin cancer patients are treated in United States, mostly with nonmelanoma skin cancer (NMSC), which is also frequent in other Western countries. As sunscreens do not provide adequate protection against deleterious effects of UVR, additional and alternative chemoprevention strategies are urgently needed to reduce skin cancer burden. Over the last couple of decades, extensive research has been conducted to understand the molecular basis of skin carcinogenesis, and to identifying novel agents which could be useful in the chemoprevention of skin cancer. In this regard, several natural non-toxic compounds have shown promising efficacy in preventing skin carcinogenesis at initiation, promotion and progression stages, and are considered important in better management of skin cancer. Consistent with this, we and others have studied and established the notable efficacy of natural flavonolignan silibinin against UVB-induced skin carcinogenesis. Extensive pre-clinical animal and cell culture studies report strong anti-inflammatory, anti-oxidant, DNA damage repair, immune-modulatory and anti-proliferative properties of silibinin. Molecular studies have identified that silibinin targets pleotropic signaling pathways including mitogenic, cell cycle, apoptosis, autophagy, p53, NF-κB, etc. Overall, the skin cancer chemopreventive potential of silibinin is well supported by comprehensive mechanistic studies, suggesting its greater use against UV-induced cellular damages and photocarcinogenesis.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences ; University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences ; University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado 80045, USA
| |
Collapse
|
256
|
|
257
|
ARHI overexpression induces epithelial ovarian cancer cell apoptosis and excessive autophagy. Int J Gynecol Cancer 2015; 24:437-43. [PMID: 24476894 DOI: 10.1097/igc.0000000000000065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE ARHI is a maternally imprinted tumor suppressor gene that is responsible for initiating programmed cell death and inhibiting cancer cell growth. However, the influence of ARHI on epithelial ovarian cancer cell death and the underlying mechanisms behind how ARHI regulates cancer cells still require further studies. METHODS Epithelial ovarian cancer cells TOV112D and ES-2 were used in this in vitro study. Cell proliferation, apoptosis, and autophagy activities were compared in TOV112D and ES-2 cells transfected with ARHI vectors or control vectors. Bcl-2 siRNA was transfected into TOV112D cells to investigate the roles of Bcl-2 played in regulating apoptosis and autophagy. RESULTS ARHI expression was reduced in TOV112D and ES-2 cells compared with normal epithelial ovarian cells (NOE095 and HOSEpiC). Overexpressed ARHI inhibited cancer cell proliferation, whereas induced forced cell apoptosis and excessive formation of autophagosomes inhibited promoted cell death. Furthermore, we found that Bcl-2 expression moderately declined in response to ARHI overexpressing in ES-2 and TOV112D cells; meanwhile, more apoptotic cells and higher LC3 level presented after silence of Bcl-2 in TOV112D cells. Reduced Bcl-2-Beclin 1 complex were observed in ARHI overexpressing cells. Moreover, modulation of ARHI to Bcl-2 expression could be ascribed partially to the activation of PI3k/AKT pathway. The addition of LY294002 enabled to suppress Bcl-2 expression and cell proliferation. CONCLUSIONS The silence of ARHI expression in vitro seems to accelerate the malignant transformation of healthy ovarian cells by restraining apoptosis and autophagy. The overexpressed ARHI in TOV112D cancer cells suppresses the activation of PI3K/AKT and reduces the expression of Bcl-2, leading to enhanced cell apoptosis and autophagic cancer cell death.
Collapse
|
258
|
In vitro and in vivo antimutagenic effects of DIG, a herbal preparation of Berberis vulgaris, Taraxacum officinale and Arctium lappa, against mitomycin C. J Nat Med 2015; 69:267-77. [DOI: 10.1007/s11418-015-0886-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/15/2015] [Indexed: 01/07/2023]
|
259
|
Peroxiredoxin 1 promotes tumorigenesis through regulating the activity of mTOR/p70S6K pathway in esophageal squamous cell carcinoma. Med Oncol 2015; 32:455. [PMID: 25579166 DOI: 10.1007/s12032-014-0455-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/12/2014] [Indexed: 01/16/2023]
Abstract
The biological function of Peroxiredoxin 1 (Prdx1) in cancer is still ambiguous, and its mechanism has not been elucidated so far. Previous studies have shown that Prdx1 functions as tumor suppressor in several types of cancers, but other studies have indicated that it is overexpressed in some types of human cancers, and inhibition of Prdx1 by shRNA contributes to radiosensitivity and chemosensitivity. In this study, a suppression subtractive hybridization cDNA library between esophageal squamous cell carcinoma (ESCC) cell line EC9706 and noncancerous esophageal epithelial cell line Het-1A was constructed, and 11 tumorigenesis-associated genes including Prdx1 were isolated. In addition, we further confirmed that Prdx1 was overexpressed in ESCC cells at the level of protein compared with Het-1A (P < 0.05). Inhibition of Prdx1 by shRNA lentivirus decreased cell proliferation and invasion, and induced cell apoptosis, but did not affect cell cycle distribution of EC9706 cells (P > 0.05). Importantly, the total proteins of mTOR and p70S6K, as well as the activity of mTOR/p70S6K signaling pathway, were decreased in Prdx1-depletion EC9706 cells. Furthermore, the activity of mTOR/p70S6K signaling pathway was increased in Prdx1-overexpressing Het-1A cells. These findings mentioned above demonstrate that Prdx1 may be involved in tumorigenesis through regulation of mTOR/p70S6K pathway in ESCC.
Collapse
|
260
|
Inhibition of oxidative metabolism leads to p53 genetic inactivation and transformation in neural stem cells. Proc Natl Acad Sci U S A 2015; 112:1059-64. [PMID: 25583481 DOI: 10.1073/pnas.1413165112] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alterations of mitochondrial metabolism and genomic instability have been implicated in tumorigenesis in multiple tissues. High-grade glioma (HGG), one of the most lethal human neoplasms, displays genetic modifications of Krebs cycle components as well as electron transport chain (ETC) alterations. Furthermore, the p53 tumor suppressor, which has emerged as a key regulator of mitochondrial respiration at the expense of glycolysis, is genetically inactivated in a large proportion of HGG cases. Therefore, it is becoming evident that genetic modifications can affect cell metabolism in HGG; however, it is currently unclear whether mitochondrial metabolism alterations could vice versa promote genomic instability as a mechanism for neoplastic transformation. Here, we show that, in neural progenitor/stem cells (NPCs), which can act as HGG cell of origin, inhibition of mitochondrial metabolism leads to p53 genetic inactivation. Impairment of respiration via inhibition of complex I or decreased mitochondrial DNA copy number leads to p53 genetic loss and a glycolytic switch. p53 genetic inactivation in ETC-impaired neural stem cells is caused by increased reactive oxygen species and associated oxidative DNA damage. ETC-impaired cells display a marked growth advantage in the presence or absence of oncogenic RAS, and form undifferentiated tumors when transplanted into the mouse brain. Finally, p53 mutations correlated with alterations in ETC subunit composition and activity in primary glioma-initiating neural stem cells. Together, these findings provide previously unidentified insights into the relationship between mitochondria, genomic stability, and tumor suppressive control, with implications for our understanding of brain cancer pathogenesis.
Collapse
|
261
|
Layman WS, Zuo J. Epigenetic regulation in the inner ear and its potential roles in development, protection, and regeneration. Front Cell Neurosci 2015; 8:446. [PMID: 25750614 PMCID: PMC4285911 DOI: 10.3389/fncel.2014.00446] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/11/2014] [Indexed: 11/13/2022] Open
Abstract
The burgeoning field of epigenetics is beginning to make a significant impact on our understanding of tissue development, maintenance, and function. Epigenetic mechanisms regulate the structure and activity of the genome in response to intracellular and environmental cues that direct cell-type specific gene networks. The inner ear is comprised of highly specialized cell types with identical genomes that originate from a single totipotent zygote. During inner ear development specific combinations of transcription factors and epigenetic modifiers must function in a coordinated manner to establish and maintain cellular identity. These epigenetic regulatory mechanisms contribute to the maintenance of distinct chromatin states and cell-type specific gene expression patterns. In this review, we highlight emerging paradigms for epigenetic modifications related to inner ear development, and how epigenetics may have a significant role in hearing loss, protection, and regeneration.
Collapse
Affiliation(s)
- Wanda S Layman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital Memphis, TN, USA
| | - Jian Zuo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital Memphis, TN, USA
| |
Collapse
|
262
|
Sridharan DM, Asaithamby A, Bailey SM, Costes SV, Doetsch PW, Dynan WS, Kronenberg A, Rithidech KN, Saha J, Snijders AM, Werner E, Wiese C, Cucinotta FA, Pluth JM. Understanding cancer development processes after HZE-particle exposure: roles of ROS, DNA damage repair and inflammation. Radiat Res 2015; 183:1-26. [PMID: 25564719 DOI: 10.1667/rr13804.1] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
During space travel astronauts are exposed to a variety of radiations, including galactic cosmic rays composed of high-energy protons and high-energy charged (HZE) nuclei, and solar particle events containing low- to medium-energy protons. Risks from these exposures include carcinogenesis, central nervous system damage and degenerative tissue effects. Currently, career radiation limits are based on estimates of fatal cancer risks calculated using a model that incorporates human epidemiological data from exposed populations, estimates of relative biological effectiveness and dose-response data from relevant mammalian experimental models. A major goal of space radiation risk assessment is to link mechanistic data from biological studies at NASA Space Radiation Laboratory and other particle accelerators with risk models. Early phenotypes of HZE exposure, such as the induction of reactive oxygen species, DNA damage signaling and inflammation, are sensitive to HZE damage complexity. This review summarizes our current understanding of critical areas within the DNA damage and oxidative stress arena and provides insight into their mechanistic interdependence and their usefulness in accurately modeling cancer and other risks in astronauts exposed to space radiation. Our ultimate goals are to examine potential links and crosstalk between early response modules activated by charged particle exposure, to identify critical areas that require further research and to use these data to reduced uncertainties in modeling cancer risk for astronauts. A clearer understanding of the links between early mechanistic aspects of high-LET response and later surrogate cancer end points could reveal key nodes that can be therapeutically targeted to mitigate the health effects from charged particle exposures.
Collapse
Affiliation(s)
- D M Sridharan
- a Lawrence Berkeley National Laboratory, Berkeley, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Nrf2, the master redox switch: The Achilles' heel of ovarian cancer? Biochim Biophys Acta Rev Cancer 2014; 1846:494-509. [DOI: 10.1016/j.bbcan.2014.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/13/2014] [Accepted: 09/17/2014] [Indexed: 12/21/2022]
|
264
|
Fu Y, He CJ, Ji PY, Zhuo ZY, Tian XZ, Wang F, Tan DX, Liu GS. Effects of melatonin on the proliferation and apoptosis of sheep granulosa cells under thermal stress. Int J Mol Sci 2014; 15:21090-104. [PMID: 25405739 PMCID: PMC4264214 DOI: 10.3390/ijms151121090] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 12/27/2022] Open
Abstract
The cross-talk between oocyte and somatic cells plays a crucial role in the regulation of follicular development and oocyte maturation. As a result, granulosa cell apoptosis causes follicular atresia. In this study, sheep granulosa cells were cultured under thermal stress to induce apoptosis, and melatonin (MT) was examined to evaluate its potential effects on heat-induced granulosa cell injury. The results demonstrated that the Colony Forming Efficiency (CFE) of granulosa cells was significantly decreased (heat 19.70% ± 1.29% vs. control 26.96% ± 1.81%, p < 0.05) and the apoptosis rate was significantly increased (heat 56.16% ± 13.95%vs. control 22.80% ± 12.16%, p < 0.05) in granulosa cells with thermal stress compared with the control group. Melatonin (10−7 M) remarkably reduced the negative effects caused by thermal stress in the granulosa cells. This reduction was indicated by the improved CFE and decreased apoptotic rate of these cells. The beneficial effects of melatonin on thermal stressed granulosa cells were not inhibited by its membrane receptor antagonist luzindole. A mechanistic exploration indicated that melatonin (10−7 M) down-regulated p53 and up-regulated Bcl-2 and LHR gene expression of granulosa cells under thermal stress. This study provides evidence for the molecular mechanisms of the protective effects of melatonin on granulosa cells during thermal stress.
Collapse
Affiliation(s)
- Yao Fu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Chang-Jiu He
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Peng-Yun Ji
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Zhi-Yong Zhuo
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Xiu-Zhi Tian
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Feng Wang
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Dun-Xian Tan
- Department of Cellular & Structural Biology, The UT Health Science Center, San Antonio, TX 78229, USA.
| | - Guo-Shi Liu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, National Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
265
|
Valencia Antúnez CA, Taja Chayeb L, Rodríguez-Segura MÁ, López Álvarez GS, García-Cuéllar CM, Villa Treviño S. DNA methyltransferases 3a and 3b are differentially expressed in the early stages of a rat liver carcinogenesis model. Oncol Rep 2014; 32:2093-103. [PMID: 25190601 DOI: 10.3892/or.2014.3462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/23/2014] [Indexed: 11/06/2022] Open
Abstract
Carcinogenesis is driven by the accumulation of mutations and abnormal DNA methylation patterns, particularly the hypermethylation of tumor‑suppressor genes. Changes in genomic DNA methylation patterns are established by the DNA methyltransferases (DNMTs) family: DNMT1, DNMT3a and DNMT3b. The DNMTs are known to be overexpressed in tumors. However, when the DNMTs expression profile is altered in earlier stages of carcinogenesis remains to be elucidated. The resistant hepatocyte model (RHM) allows the analysis of the hepatocellular carcinoma (HCC) from the formation of altered cell foci to the appearance of tumors in rats. To investigate the DNMTs expression in this model, we first observed that timp3, rassf1a and p16 genes became methylated during cancer development by methylation‑specific PCR (MSP) and the bisulphate sequencing PCR (BSP) of timp3. The differential expression at the RNA and protein level of the three DNMTs was also assessed. dnmt1 expression was higher in tumors than in normal and early cancer stages. However, no evident overexpression of the enzyme was identified by immunohistochemistry. By contrast, DNMT3a and DNMT3b were consistently subexpressed in tumors. In the present study, we report a carcinogenesis model that does not feature the overexpression of DNMT1 but exhibits a transient expression of DNMT3a and DNMT3b.
Collapse
Affiliation(s)
- Carlos Alberto Valencia Antúnez
- Department of Cell Biology Center for Research and Advanced Studies (CINVESTAV) IPN, Basic Research Branch, Mexico, D.F., Mexico
| | - Lucía Taja Chayeb
- National Cancer Institute, Basic Research Branch, Mexico, D.F., Mexico
| | - Miguel Ángel Rodríguez-Segura
- Department of Physics, Center for Research and Advanced Studies (CINVESTAV) IPN, Basic Research Branch, Mexico, D.F., Mexico
| | - Guadalupe Soledad López Álvarez
- Department of Cell Biology Center for Research and Advanced Studies (CINVESTAV) IPN, Basic Research Branch, Mexico, D.F., Mexico
| | | | - Saúl Villa Treviño
- Department of Cell Biology Center for Research and Advanced Studies (CINVESTAV) IPN, Basic Research Branch, Mexico, D.F., Mexico
| |
Collapse
|
266
|
Sex-specific dose-response analysis of genotoxicity in cyproterone acetate-treated F344 rats. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 774:1-7. [DOI: 10.1016/j.mrgentox.2014.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/18/2014] [Accepted: 08/23/2014] [Indexed: 11/19/2022]
|
267
|
Zorofchian Moghadamtousi S, Karimian H, Rouhollahi E, Paydar M, Fadaeinasab M, Abdul Kadir H. Annona muricata leaves induce G₁ cell cycle arrest and apoptosis through mitochondria-mediated pathway in human HCT-116 and HT-29 colon cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2014; 156:277-289. [PMID: 25195082 DOI: 10.1016/j.jep.2014.08.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 07/19/2014] [Accepted: 08/11/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Annona muricata known as "the cancer killer" has been widely used in the traditional medicine for the treatment of cancer and tumors. The purpose of this study is to investigate the anticancer properties of ethyl acetate extract of Annona muricata leaves (EEAM) on HT-29 and HCT-116 colon cancer cells and the underlying mechanisms. MATERIALS AND METHODS The effect of EEAM on the cell proliferation of HT-29 and HCT-116 cells was analyzed by the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium) assay. High content screening system (HCS) was applied to investigate the cell membrane permeability, mitochondrial membrane potential (MMP), nuclear condensation and cytochrome c translocation from mitochondria to cytosol. Reactive oxygen species (ROS) formation, lactate dehydrogenase (LDH) release and activation of caspase-3/7, -8 and -9 were measured while treatment. Flow cytometric analysis was used to determine the cell cycle distribution and phosphatidylserine externalization. The protein expression of Bax and Bcl-2 was determined using immunofluorescence analysis. In addition, the potential of EEAM to suppress the migration and invasion of colon cancer cells was also examined. RESULTS EEAM exerted significant cytotoxic effects on HCT-116 and HT-29 cells as determined by MTT and LDH assays. After 24 h treatment, EEAM exhibited the IC₅₀ value of 11.43 ± 1.87 µg/ml and 8.98 ± 1.24 µg/ml against HT-29 and HCT-116 cells, respectively. Flow cytometric analysis demonstrated the cell cycle arrest at G1 phase and phosphatidylserine externalization confirming the induction of apoptosis. EEAM treatment caused excessive accumulation of ROS followed by disruption of MMP, cytochrome c leakage and activation of the initiator and executioner caspases in both colon cancer cells. Immunofluorescence analysis depicted the up-regulation of Bax and down-regulation of Bcl-2 proteins while treated with EEAM. Furthermore, EEAM conspicuously blocked the migration and invasion of HT-29 and HCT-116 cells. CONCLUSIONS These findings provide a scientific basis for the use of A. muricata leaves in the treatment of cancer, although further in vivo studies are still required.
Collapse
Affiliation(s)
- Soheil Zorofchian Moghadamtousi
- Biomolecular Research Group, Biochemistry Program, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Elham Rouhollahi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohammadjavad Paydar
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mehran Fadaeinasab
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Habsah Abdul Kadir
- Biomolecular Research Group, Biochemistry Program, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
268
|
Chen Y, Williams V, Filippova M, Filippov V, Duerksen-Hughes P. Viral carcinogenesis: factors inducing DNA damage and virus integration. Cancers (Basel) 2014; 6:2155-86. [PMID: 25340830 PMCID: PMC4276961 DOI: 10.3390/cancers6042155] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/03/2014] [Accepted: 10/09/2014] [Indexed: 12/13/2022] Open
Abstract
Viruses are the causative agents of 10%-15% of human cancers worldwide. The most common outcome for virus-induced reprogramming is genomic instability, including accumulation of mutations, aberrations and DNA damage. Although each virus has its own specific mechanism for promoting carcinogenesis, the majority of DNA oncogenic viruses encode oncogenes that transform infected cells, frequently by targeting p53 and pRB. In addition, integration of viral DNA into the human genome can also play an important role in promoting tumor development for several viruses, including HBV and HPV. Because viral integration requires the breakage of both the viral and the host DNA, the integration rate is believed to be linked to the levels of DNA damage. DNA damage can be caused by both endogenous and exogenous factors, including inflammation induced by either the virus itself or by co-infections with other agents, environmental agents and other factors. Typically, cancer develops years to decades following the initial infection. A better understanding of virus-mediated carcinogenesis, the networking of pathways involved in transformation and the relevant risk factors, particularly in those cases where tumorigenesis proceeds by way of virus integration, will help to suggest prophylactic and therapeutic strategies to reduce the risk of virus-mediated cancer.
Collapse
Affiliation(s)
- Yan Chen
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Vonetta Williams
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Maria Filippova
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Valery Filippov
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | | |
Collapse
|
269
|
Bak MJ, Jeong WS, Kim KB. Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells. Int J Mol Med 2014; 34:1516-22. [PMID: 25319719 DOI: 10.3892/ijmm.2014.1972] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/08/2014] [Indexed: 11/06/2022] Open
Abstract
Fermented black ginseng (FBG) is prepared by repeated steaming and drying processes with fresh ginseng followed by fermentation with Saccharomyces cerevisiae. It has recently been shown to have several bioactivities. FBG contains crude saponin (1,440 µg/ml), ginsenoside Rg2 (2.86 µg/ml), ginsenoside Rg3 (24.52 µg/ml), ginsenoside Rh1 (12.64 µg/ml), ginsenoside Rh2 (0.63 µg/ml) and ginsenoside Rf (1.32 µg/ml). The present study investigated the antioxidant defense properties of FBG against hydrogen peroxide (H2O2)-mediated oxidative stress in HepG2 human hepatocellular carcinoma cells. The increased production of reactive oxygen species (ROS) induced by H2O2 was attenuated in a dose-dependent manner when the cells were pre-treated with FBG (10-50 µg/ml). FBG induced both the expression and activity of antioxidant enzymes, such as superoxide dismutase, catalase and glutathione peroxidase in the H2O2-treated HepG2 cells. The inhibitory effects of FBG on the phosphorylation of upstream mitogen-activated protein kinases (MAPKs), such as c-Jun N-terminal kinase, extracellular signal-regulated kinase and p38 were also observed. Overall, our results demonstrate that FBG protects HepG2 cells from oxidative stress through the induction of antioxidant enzyme activity and the inhibition of MAPK pathways.
Collapse
Affiliation(s)
- Min-Ji Bak
- Department of Food and Life Sciences, College of Biomedical Science and Engineering, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Woo-Sik Jeong
- Department of Food and Life Sciences, College of Biomedical Science and Engineering, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Kyu-Bong Kim
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungnam 330-714, Republic of Korea
| |
Collapse
|
270
|
Kim TH, Song J, Kim SH, Parikh AK, Mo X, Palanichamy K, Kaur B, Yu J, Yoon SO, Nakano I, Kwon CH. Piperlongumine treatment inactivates peroxiredoxin 4, exacerbates endoplasmic reticulum stress, and preferentially kills high-grade glioma cells. Neuro Oncol 2014; 16:1354-64. [PMID: 24879047 PMCID: PMC4165421 DOI: 10.1093/neuonc/nou088] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 04/12/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDS Piperlongumine, a natural plant product, kills multiple cancer types with little effect on normal cells. Piperlongumine raises intracellular levels of reactive oxygen species (ROS), a phenomenon that may underlie the cancer-cell killing. Although these findings suggest that piperlongumine could be useful for treating cancers, the mechanism by which the drug selectively kills cancer cells remains unknown. METHODS We treated multiple high-grade glioma (HGG) sphere cultures with piperlongumine and assessed its effects on ROS and cell-growth levels as well as changes in downstream signaling. We also examined the levels of putative piperlongumine targets and their roles in HGG cell growth. RESULTS Piperlongumine treatment increased ROS levels and preferentially killed HGG cells with little effect in normal brain cells. Piperlongumine reportedly increases ROS levels after interactions with several redox regulators. We found that HGG cells expressed higher levels of the putative piperlongumine targets than did normal neural stem cells (NSCs). Furthermore, piperlongumine treatment in HGG cells, but not in normal NSCs, increased oxidative inactivation of peroxiredoxin 4 (PRDX4), an ROS-reducing enzyme that is overexpressed in HGGs and facilitates proper protein folding in the endoplasmic reticulum (ER). Moreover, piperlongumine exacerbated intracellular ER stress, an effect that was mimicked by suppressing PRDX4 expression. CONCLUSIONS Our results reveal that the mechanism by which piperlongumine preferentially kills HGG cells involves PRDX4 inactivation, thereby inducing ER stress. Therefore, piperlongumine treatment could be considered as a novel therapeutic option for HGG treatment.
Collapse
Affiliation(s)
- Tae Hyong Kim
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Jieun Song
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Sung-Hak Kim
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Arav Krishnavadan Parikh
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Xiaokui Mo
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Kamalakannan Palanichamy
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Balveen Kaur
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Jianhua Yu
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Sung Ok Yoon
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Ichiro Nakano
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| | - Chang-Hyuk Kwon
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, Ohio State University, Columbus, Ohio (T.H.K., J.S., S.-H.K., A.K.P., I.N., B.K., C.-H.K.); Solid Tumor Program at the James Comprehensive Cancer Center, Columbus, Ohio (T.H.K., J.S., A.K.P., C.-H.K.); Center for Biostatistics, Ohio State University, Columbus, Ohio (X.M.); Department of Radiation Oncology, Ohio State University, Columbus, Ohio (K.P.); Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio (J.Y.); Department of Molecular and Cellular Biochemistry, Ohio State University Wexner Medical Center, Columbus, Ohio (S.O.Y.)
| |
Collapse
|
271
|
Roca-Rodríguez MDM, García-Almeida JM, Ruiz-Nava J, Alcaide J, Lupiañez-Pérez Y, Rico-Pérez JM, Toledo-Serrano MD, Cardona F, Medina-Carmona JA, Tinahones FJ. Long-term effects of varying consumption of ω3 fatty acids in ear, nose and throat cancer patients: assessment 1 year after radiotherapy. Int J Food Sci Nutr 2014; 66:108-13. [PMID: 25265206 DOI: 10.3109/09637486.2014.953453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A prospective 1-year follow-up study in ear, nose, and throat (ENT) cancer patients was carried out one year after radiotherapy to assess the effect of varying consumption of ω3 fatty acid according to whether they consumed more or less than the 50th percentile of ω3 fatty acids. Clinical, analytical, inflammatory (CRP and IL-6), and oxidative variables (TAC, GPx, GST, and SOD) were evaluated. The study comprised 31 patients (87.1% men), with a mean age of 61.3 ± 9.1 years. Hematological variables showed significant differences in the patients with a lower consumption of ω3 fatty acids. A lower mortality and longer survival were found in the group with ω3 fatty acid consumption ≥50th percentile but the differences were not significant. No significant difference was reached in toxicity, inflammation, and oxidative stress markers. The group with ω3 fatty acid consumption <50th percentile significantly experienced more hematological and immune changes.
Collapse
Affiliation(s)
- María Del Mar Roca-Rodríguez
- Biomedical Research Institute of Malaga (IBIMA), Virgen de la Victoria Hospital, University of Malaga , Málaga , Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
272
|
Anestopoulos I, Voulgaridou GP, Georgakilas AG, Franco R, Pappa A, Panayiotidis MI. Epigenetic therapy as a novel approach in hepatocellular carcinoma. Pharmacol Ther 2014; 145:103-19. [PMID: 25205159 DOI: 10.1016/j.pharmthera.2014.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/02/2014] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver malignancy and one with high fatality. Its 5-year survival rate remains low and thus, there is a need for improvement of current treatment strategies as well as development of novel targeted methodologies in order to optimize existing therapeutic protocols. To this end, only recently, it was discovered that its pathophysiology also involves epigenetic alterations in DNA methylation, histone modifications and/or non-coding microRNA patterns. Unlike genetic events, epigenetic alterations are reversible and thus potentially considered to be an alternative option in cancer treatment protocols. In this review, we describe the general characteristics and resulted major alterations of the epigenetic machinery as well as current state of progress of epigenetic therapy (via different single or combinatorial experimental approaches) in HCC.
Collapse
Affiliation(s)
- Ioannis Anestopoulos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Alexandros G Georgakilas
- School of Applied Mathematical & Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Rodrigo Franco
- Redox Biology Center, School of Veterinary Medicine & Biomedical Sciences, Redox Biology Center, University of Nebraska-Lincoln, USA
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|
273
|
Ogunbolude Y, Ibrahim M, Elekofehinti OO, Adeniran A, Abolaji AO, Rocha JBT, Kamdem JP. Effects of Tapinanthus globiferus and Zanthoxylum zanthoxyloides extracts on human leukocytes in vitro. JOURNAL OF COMPLEMENTARY MEDICINE RESEARCH 2014; 3:167-72. [PMID: 26401368 PMCID: PMC4576816 DOI: 10.5455/jice.20140826110059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 08/26/2014] [Indexed: 12/21/2022]
Abstract
Objective: This study aimed at investigating the genotoxicity and cytotoxicity effect of Tapinanthus globiferus and Zanthoxylum zanthoxyloides to human leukocytes. In addition, the reductive potential and the chemical composition of the two plant extracts were also determined. Materials and Methods: Human leukocytes were obtained from healthy volunteer donors. The genotoxicity and cytotoxicity of T. globiferus and Z. zanthoxyloides were assessed using the comet assay and trypan blue exclusion, respectively. The antioxidant activity of the plant extracts was evaluated by the reducing power assay. Furthermore, high-performance liquid chromatography-diode array detector was used to characterize and quantify the constituents of these plants. Results: T. globiferus (10-150 µg/mL) was neither genotoxic nor cytotoxic at the concentrations tested, suggesting that it can be consumed safely at relatively high concentrations. However, Z. zanthoxyloides showed cytoxicity and genotoxicity to human leukocytes at the highest concentration tested (150 µg/mL). In addition, the total reducing power of T. globiferus was found higher than Z. zanthoxyloides in potassium ferricyanide reduction. Both plants extract contained flavonoids (rutin and quercetin) and phenolic acids (chlorogenic and caffeic). Conclusion: The results obtained support the fact that some caution should be paid regarding the dosage and the frequency of use of Z. zanthoxyloides extract.
Collapse
Affiliation(s)
- Yetunde Ogunbolude
- Department of Biochemistry, Federal University of Technology, PMB 704, Akure, Ondo, Nigeria
| | - Mohammad Ibrahim
- Department of Chemistry, Abdul Wali Khan University, Mardan Pakistan
| | | | - Adekunle Adeniran
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Amos Olalekan Abolaji
- Department of Biochemistry, Drug Metabolism and Molecular Toxicology Research Laboratories, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - João Batista Teixeira Rocha
- Department of Biochemistry and Molecular Biology, Post-Graduate Program in Biochemical Toxicology, Federal University of Santa Maria, Santa Maria-RS, CEP 97105-900, Brazil
| | - Jean Paul Kamdem
- Department of Biochemistry and Molecular Biology, Post-Graduate Program in Biochemical Toxicology, Federal University of Santa Maria, Santa Maria-RS, CEP 97105-900, Brazil ; Department of Biochemistry, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, CEP 90035-003, RS, Brazil
| |
Collapse
|
274
|
El Hosry L, Di Giorgio C, Birer C, Habib J, Tueni M, Bun SS, Herbette G, De Meo M, Ollivier E, Elias R. In vitro cytotoxic and anticlastogenic activities of saxifragifolin B and cyclamin isolated from Cyclamen persicum and Cyclamen libanoticum. PHARMACEUTICAL BIOLOGY 2014; 52:1134-1140. [PMID: 24649909 DOI: 10.3109/13880209.2013.879600] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT The genus Cyclamen L. (Primulaceae) is rich in saponins known to have interesting biological activities. OBJECTIVE To isolate saxifragifolin B and cyclamin, two triterpene saponins, from Cyclamen libanoticum Hildebr and Cyclamen persicum Mill, and to assess their cytotoxic, clastogenic/aneugenic, and anticlastogenic effects, as well as antioxidant potential. MATERIALS AND METHODS Saxifragifolin B and cyclamin were tested for their cytotoxicity against SK-BR-3, HT-29, HepG2/3A, NCI-H1299, BXPC-3, 22RV1, and normal DMEM cell lines using WST-1 assay. Their clastogenic/aneugenic activities and anticlastogenic effects against the anticancer drug mitomycin C were assessed by the in vitro micronucleus assay in CHO cells. Their antioxidant capacities were determined using Fe(2+)-chelating and 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging assays. RESULTS Both saponins were described for the first time in Cyclamen libanoticum. They showed strong cytotoxic activities against the tested cancer cell lines. Saxifragifolin B was found to be 56- and 37-times more active than mitomycin C against breast adenocarcinoma (SK-BR-3) and lung carcinoma (NCI-H1299), respectively. Also, saxifragifolin B did not induce micronuclei formation and prevented cells from mitomycin C clastogenic effect. Cyclamin induced a significant increase of micronucleated cells after metabolic activation with S9 mix, and did not possess any anticlastogenic activity. Both molecules exhibited low antioxidant activities as compared to reference compounds. DISCUSSION AND CONCLUSIONS This study showed the remarkable cytotoxic activity of saxifragifolin B, especially against breast adenocarcinoma and lung carcinoma and its chemoprotective activity against mitomycin C. Thus, saxifragifolin B could be suggested as a potential cytotoxic drug with a preventive effect against possible exposures to genotoxic agents.
Collapse
Affiliation(s)
- Leina El Hosry
- Laboratoire de Pharmacognosie et Ethnopharmacologie, UMR MD3, Faculté de Pharmacie, Aix-Marseille Université , Marseille Cedex , France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
275
|
ETV6/RUNX1 induces reactive oxygen species and drives the accumulation of DNA damage in B cells. Neoplasia 2014; 15:1292-300. [PMID: 24339741 DOI: 10.1593/neo.131310] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/21/2022] Open
Abstract
The t(12;21)(p13;q22) chromosomal translocation is the most frequent translocation in childhood B cell precursor-acute lymphoblastic leukemia and results in the expression of an ETV6/RUNX1 fusion protein. The frequency of ETV6/RUNX1 fusions in newborns clearly exceeds the leukemia rate revealing that additional events occur in ETV6/RUNX1-positive cells for leukemic transformation. Hitherto, the mechanisms triggering these second hits remain largely elusive. Thus, we generated a novel ETV6/RUNX1 transgenic mouse model where the expression of the fusion protein is restricted to CD19(+) B cells. These animals harbor regular B cell development and lack gross abnormalities. We established stable pro-B cell lines carrying the ETV6/RUNX1 transgene that allowed us to investigate whether ETV6/RUNX1 itself favors the acquisition of second hits. Remarkably, these pro-B cell lines as well as primary bone marrow cells derived from ETV6/RUNX1 transgenic animals display elevated levels of reactive oxygen species (ROS) as tested with ETV6/RUNX1 transgenic dihydroethidium staining. In line, intracellular phospho-histone H2AX flow cytometry and comet assay revealed increased DNA damage indicating that ETV6/RUNX1 expression enhances ROS. On the basis of our data, we propose the following model: the expression of ETV6/RUNX1 creates a preleukemic clone and leads to increased ROS levels. These elevated ROS favor the accumulation of secondary hits by increasing genetic instability and double-strand breaks, thus allowing preleukemic clones to develop into fully transformed leukemic cells.
Collapse
|
276
|
Enterococcus faecalis Infection and Reactive Oxygen Species Down-Regulates the miR-17-92 Cluster in Gastric Adenocarcinoma Cell Culture. Genes (Basel) 2014; 5:726-38. [PMID: 25170597 PMCID: PMC4198927 DOI: 10.3390/genes5030726] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/14/2014] [Accepted: 08/13/2014] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation due to bacterial overgrowth of the stomach predisposes to the development of gastric cancer and is also associated with high levels of reactive oxygen species (ROS). In recent years increasing attention has been drawn to microRNAs (miRNAs) due to their role in the pathogenesis of many human diseases including gastric cancer. Here we studied the impact of infection by the gram-positive bacteria Enterococcus faecalis (E. faecalis) on global miRNA expression as well as the effect of ROS on selected miRNAs. Human gastric adenocarcinoma cell line MKN74 was infected with living E. faecalis for 24 h or for 5 days or with E. faecalis lysate for 5 days. The miRNA expression was examined by microarray analysis using Affymetrix GeneChip miRNA Arrays. To test the effect of ROS, MKN74 cells were treated with 100 mM tert-Butyl hydroperoxide (TBHP). Following 5 days of E. faecalis infection we found 91 differentially expressed miRNAs in response to living bacteria and 2 miRNAs responded to E. faecalis lysate. We verified the down-regulation of the miR-17-92 and miR-106-363 clusters and of other miRNAs involved in the oxidative stress-response by qRT-PCR. We conclude that only infection by living E. faecalis bacteria caused a significant global response in miRNA expression in the MKN74 cell culture. E. faecalis infection as well as ROS stimulation down-regulated the expression of the miR-17-92 cluster. We believe that these changes could reflect a general response of gastric epithelial cells to bacterial infections.
Collapse
|
277
|
Meseguer S, Martínez-Zamora A, García-Arumí E, Andreu AL, Armengod ME. The ROS-sensitive microRNA-9/9* controls the expression of mitochondrial tRNA-modifying enzymes and is involved in the molecular mechanism of MELAS syndrome. Hum Mol Genet 2014; 24:167-84. [PMID: 25149473 DOI: 10.1093/hmg/ddu427] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mitochondrial dysfunction activates mitochondria-to-nucleus signaling pathways whose components are mostly unknown. Identification of these components is important to understand the molecular mechanisms underlying mitochondrial diseases and to discover putative therapeutic targets. MELAS syndrome is a rare neurodegenerative disease caused by mutations in mitochondrial (mt) DNA affecting mt-tRNA(Leu(UUR)). Patient and cybrid cells exhibit elevated oxidative stress. Moreover, mutant mt-tRNAs(Leu(UUR)) lack the taurine-containing modification normally present at the wobble uridine (U34) of wild-type mt-tRNA(Leu(UUR)), which is considered an etiology of MELAS. However, the molecular mechanism is still unclear. We found that MELAS cybrids exhibit a significant decrease in the steady-state levels of several mt-tRNA-modification enzymes, which is not due to transcriptional regulation. We demonstrated that oxidative stress mediates an NFkB-dependent induction of microRNA-9/9*, which acts as a post-transcriptional negative regulator of the mt-tRNA-modification enzymes GTPBP3, MTO1 and TRMU. Down-regulation of these enzymes by microRNA-9/9* affects the U34 modification status of non-mutant tRNAs and contributes to the MELAS phenotype. Anti-microRNA-9 treatments of MELAS cybrids reverse the phenotype, whereas miR-9 transfection of wild-type cells mimics the effects of siRNA-mediated down-regulation of GTPBP3, MTO1 and TRMU. Our data represent the first evidence that an mt-DNA disease can directly affect microRNA expression. Moreover, we demonstrate that the modification status of mt-tRNAs is dynamic and that cells respond to stress by modulating the expression of mt-tRNA-modifying enzymes. microRNA-9/9* is a crucial player in mitochondria-to-nucleus signaling as it regulates expression of nuclear genes in response to changes in the functional state of mitochondria.
Collapse
Affiliation(s)
- Salvador Meseguer
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Ana Martínez-Zamora
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Elena García-Arumí
- Hospital Universitari Vall d'Hebron, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain Biomedical Research Networking Centre for Rare Diseases (CIBERER) (node U701), Barcelona, Spain and
| | - Antonio L Andreu
- Hospital Universitari Vall d'Hebron, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain Biomedical Research Networking Centre for Rare Diseases (CIBERER) (node U701), Barcelona, Spain and
| | - M-Eugenia Armengod
- Laboratory of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain CIBERER (node U721), Valencia, Spain
| |
Collapse
|
278
|
Autsavapromporn N, Plante I, Liu C, Konishi T, Usami N, Funayama T, Azzam EI, Murakami T, Suzuki M. Genetic changes in progeny of bystander human fibroblasts after microbeam irradiation with X-rays, protons or carbon ions: The relevance to cancer risk. Int J Radiat Biol 2014; 91:62-70. [DOI: 10.3109/09553002.2014.950715] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
279
|
Collado R, Ivars D, Oliver I, Tormos C, Egea M, Miguel A, Sáez GT, Carbonell F. Increased oxidative damage associated with unfavorable cytogenetic subgroups in chronic lymphocytic leukemia. BIOMED RESEARCH INTERNATIONAL 2014; 2014:686392. [PMID: 25054143 PMCID: PMC4099055 DOI: 10.1155/2014/686392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/30/2014] [Accepted: 06/11/2014] [Indexed: 12/12/2022]
Abstract
Oxidative stress contributes to genomic instability in chronic lymphocytic leukemia (CLL), but its relationship with the acquisition of specific chromosomal abnormalities is unknown. We recruited 55 untreated CLL patients and assessed 8-oxo-2'-deoxyguanosine (8-oxo-dG), glutathione, and malondialdehyde (MDA) levels, and we compared them among the cytogenetic subgroups established using fluorescence in situ hybridization (FISH). Significant increases in 8-oxo-dG and/or MDA were observed in patients with unfavorable cytogenetic aberrations (17p and 11q deletions) compared to the 13q deletion group. TP53 deletion patients exhibited a diminished DNA repair efficiency. Finally, cases with normal FISH also showed enhanced 8-oxo-dG, which could result in adverse outcomes.
Collapse
Affiliation(s)
- Rosa Collado
- Service of Hematology, CDB-University General Hospital of Valencia, Avenida Tres Cruces 2, 46014 Valencia, Spain
| | - David Ivars
- Department of Medicine, Faculty of Medicine, University of Valencia, Avenida Blasco Ibáñez 13, 46010 Valencia, Spain
| | - Isabel Oliver
- Department of Medicine, Faculty of Medicine, University of Valencia, Avenida Blasco Ibáñez 13, 46010 Valencia, Spain
| | - Carmen Tormos
- CIBERobn, Biomedical Network Research Centre in Physiopathology of Obesity and Nutrition, Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Mercedes Egea
- Service of Hematology, CDB-University General Hospital of Valencia, Avenida Tres Cruces 2, 46014 Valencia, Spain
| | - Amparo Miguel
- Service of Hematology, CDB-University General Hospital of Valencia, Avenida Tres Cruces 2, 46014 Valencia, Spain
| | - Guillermo T. Sáez
- CIBERobn, Biomedical Network Research Centre in Physiopathology of Obesity and Nutrition, Choupana s/n, 15706 Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Valencia, Avenida Blasco Ibáñez 13, 46010 Valencia, Spain
- Service of Clinical Analyses, CDB-University General Hospital of Valencia, Avenida Tres Cruces 2, 46014 Valencia, Spain
| | - Félix Carbonell
- Service of Hematology, CDB-University General Hospital of Valencia, Avenida Tres Cruces 2, 46014 Valencia, Spain
- Department of Medicine, Faculty of Medicine, University of Valencia, Avenida Blasco Ibáñez 13, 46010 Valencia, Spain
| |
Collapse
|
280
|
Kilic N, Yavuz Taslipinar M, Guney Y, Tekin E, Onuk E. An investigation into the serum thioredoxin, superoxide dismutase, malondialdehyde, and advanced oxidation protein products in patients with breast cancer. Ann Surg Oncol 2014; 21:4139-43. [PMID: 24962940 DOI: 10.1245/s10434-014-3859-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Reactive oxygen species (free radicals) play an important role in carcinogenesis. Extensive antioxidant defense mechanisms counteract free radicals in mammalian cells. Oxidative stress is a disturbance in the balance between the production of free radicals and antioxidant defenses. There is direct evidence that oxidative stress and lipid peroxidation (LPO) are linked to the etiology of breast cancer. The increasing global incidence of breast cancer emphasizes the need to understand the various mechanisms involved in breast tumorigenesis. The present study was undertaken to investigate the oxidative stress and antioxidant status in the blood samples of patients with breast cancer. METHODS The present study was based on 23 women who were surgically treated at Gazi University, Faculty of Medicine, Department of General Surgery. The malondialdehyde (MDA) levels as an index of LPO along with the examination of superoxide dismutase (SOD) activities and advanced oxidation protein product (AOPP) and thioredoxin (Trx) levels were determined in the blood samples of 23 patients with breast cancer and 13 healthy controls. RESULTS MDA, AOPP, and Trx levels and SOD activities were significantly higher in patients with breast cancer than the controls. CONCLUSIONS The results showed that oxidative stress may be related to breast cancer and especially some molecules, such as Trx and AOPP, may be useful biomarkers in breast cancer diagnosis and treatment. More detailed knowledge related to the pathophysiology of these molecules could provide valuable information on the origin and development of malignant tumors, such as breast cancer.
Collapse
Affiliation(s)
- Nedret Kilic
- Faculty of Medicine, Department of Medical Biochemistry, Gazi University, Ankara, Turkey
| | | | | | | | | |
Collapse
|
281
|
Causes and Consequences of Age-Related Changes in DNA Methylation: A Role for ROS? BIOLOGY 2014; 3:403-25. [PMID: 24945102 PMCID: PMC4085615 DOI: 10.3390/biology3020403] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 05/28/2014] [Accepted: 05/31/2014] [Indexed: 01/15/2023]
Abstract
Recent genome-wide analysis of C-phosphate-G (CpG) sites has shown that the DNA methylome changes with increasing age, giving rise to genome-wide hypomethylation with site‑specific incidences of hypermethylation. This notion has received a lot of attention, as it potentially explains why aged organisms generally have a higher risk of age-related diseases. However, very little is known about the mechanisms that could cause the occurrence of these changes. Moreover, there does not appear to be a clear link between popular theories of aging and alterations in the methylome. Some of the most fruitful of these theories attribute an important role to reactive oxygen species, which seem to be responsible for an increase in oxidative damage to macromolecules, such as DNA, during the lifetime of an organism. In this review, the connection between changes in DNA methylation and these reactive oxygen species is discussed, as well as the effect of these changes on health. Deeper insights into the nature, causes and consequences of the aging methylome might provide a deeper understanding of the molecular mechanisms of aging and eventually contribute to the development of new diagnostic and therapeutic tools.
Collapse
|
282
|
Cadet J, Wagner JR. Oxidatively generated base damage to cellular DNA by hydroxyl radical and one-electron oxidants: similarities and differences. Arch Biochem Biophys 2014; 557:47-54. [PMID: 24820329 DOI: 10.1016/j.abb.2014.05.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/23/2014] [Accepted: 05/01/2014] [Indexed: 01/05/2023]
Abstract
Hydroxyl radical (OH) and one-electron oxidants that may be endogenously formed through oxidative metabolism, phagocytosis, inflammation and pathological conditions constitute the main sources of oxidatively generated damage to cellular DNA. It is worth mentioning that exposure of cells to exogenous physical agents (UV light, high intensity UV laser, ionizing radiation) and chemicals may also induce oxidatively generated damage to DNA. Emphasis is placed in this short review article on the mechanistic aspects of OH and one-electron oxidant-mediated formation of single and more complex damage (tandem lesions, intra- and interstrand cross-links, DNA-protein cross-links) in cellular DNA arising from one radical hit. This concerns DNA modifications that have been accurately measured using suitable analytical methods such as high performance liquid chromatography coupled with electrospray ionization tandem mass spectrometry. Evidence is provided that OH and one-electron oxidants after generating neutral radicals and base radical cations respectively may partly induce common degradation pathways. In addition, selective oxidative reactions giving rise to specific degradation products of OH and one-electron oxidation reactions that can be used as representative biomarkers of these oxidants have been identified.
Collapse
Affiliation(s)
- Jean Cadet
- Institut Nanosciences et Cryogénie, CEA/Grenoble, F-38054 Grenoble Cedex 9, France; Département de Médecine Nucléaire et Radiobiologie, Faculté de Médecine des Sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - J Richard Wagner
- Département de Médecine Nucléaire et Radiobiologie, Faculté de Médecine des Sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
283
|
Liu-Smith F, Dellinger R, Meyskens FL. Updates of reactive oxygen species in melanoma etiology and progression. Arch Biochem Biophys 2014; 563:51-5. [PMID: 24780245 DOI: 10.1016/j.abb.2014.04.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/14/2014] [Accepted: 04/17/2014] [Indexed: 01/01/2023]
Abstract
Reactive oxygen species (ROS) play crucial roles in all aspects of melanoma development, however, the source of ROS is not well defined. In this review we summarize recent advancement in this rapidly developing field. The cellular ROS pool in melanocytes can be derived from mitochondria, melanosomes, NADPH oxidase (NOX) family enzymes, and uncoupling of nitric oxide synthase (NOS). Current evidence suggests that Nox1, Nox4 and Nox5 are expressed in melanocytic lineage. While there is no difference in Nox1 expression levels in primary and metastatic melanoma tissues, Nox4 expression is significantly higher in a subset of metastatic melanoma tumors as compared to the primary tumors; suggesting distinct and specific signals and effects for NOX family enzymes in melanoma. Targeting these NOX enzymes using specific NOX inhibitors may be effective for a subset of certain tumors. ROS also play important roles in BRAF inhibitor induced drug resistance; hence identification and blockade of the source of this ROS may be an effective way to enhance efficacy and overcome resistance. Furthermore, ROS from different sources may interact with each other and interact with reactive nitrogen species (RNS) and drive the melanomagenesis process at all stages of disease. Further understanding ROS and RNS in melanoma etiology and progression is necessary for developing new prevention and therapeutic approaches.
Collapse
Affiliation(s)
- Feng Liu-Smith
- Department of Epidemiology, University of California School of Medicine, Irvine, CA 92697, United States; Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States.
| | - Ryan Dellinger
- Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States
| | - Frank L Meyskens
- Department of Epidemiology, University of California School of Medicine, Irvine, CA 92697, United States; Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Department of Biological Chemistry, University of California School of Medicine, Irvine, CA 92697, United States; Department of Public Health, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States
| |
Collapse
|
284
|
Epigenetic modification of Nrf2 in 5-fluorouracil-resistant colon cancer cells: involvement of TET-dependent DNA demethylation. Cell Death Dis 2014; 5:e1183. [PMID: 24743738 PMCID: PMC4001304 DOI: 10.1038/cddis.2014.149] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/07/2014] [Accepted: 03/10/2014] [Indexed: 12/12/2022]
Abstract
5-Fluorouracil (5-FU) is a widely used anticancer drug for the treatment of colorectal cancer (CRC). However, resistance to 5-FU often prevents the success of chemotherapy. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a transcriptional regulator and a possible target to overcome 5-FU resistance. The present study examined epigenetic changes associated with Nrf2 induction in a human CRC cell line (SNUC5) resistant to 5-FU (SNUC5/5-FUR). Nrf2 expression, nuclear translocation, and binding to promoter were higher in SNUC5/5-FUR cells than in SNUC5 cells. The activated Nrf2 in SNUC5/5-FUR cells led to an increase in the protein expression and activity of heme oxygenase-1 (HO-1), an Nrf2-regulated gene. SNUC5/5-FUR cells produced a larger amount of reactive oxygen species (ROS) than SNUC5 cells. The siRNA- or shRNA-mediated knockdown of Nrf2 or HO-1 significantly suppressed cancer cell viability and tumor growth in vitro and in vivo, resulting in enhanced 5-FU sensitivity. Methylation-specific (MS) or real-time quantitative MS-PCR data showed hypomethylation of the Nrf2 promoter CpG islands in SNUC5/5-FUR cells compared with SNUC5 cells. Expression of the DNA demethylase ten-eleven translocation (TET) was upregulated in SNUC5/5-FUR cells. ROS generated by 5-FU upregulated TET1 expression and function, whereas antioxidant had the opposite effect. These results suggested that the mechanism underlying the acquisition of 5-FU resistance in CRC involves the upregulation of Nrf2 and HO-1 expression via epigenetic modifications of DNA demethylation.
Collapse
|
285
|
Kolybaba A, Classen AK. Sensing cellular states--signaling to chromatin pathways targeting Polycomb and Trithorax group function. Cell Tissue Res 2014; 356:477-93. [PMID: 24728925 DOI: 10.1007/s00441-014-1824-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 01/22/2014] [Indexed: 02/06/2023]
Abstract
Cells respond to extra- and intra-cellular signals by dynamically changing their gene expression patterns. After termination of the original signal, new expression patterns are maintained by epigenetic DNA and histone modifications. This represents a powerful mechanism that enables long-term phenotypic adaptation to transient signals. Adaptation of epigenetic landscapes is important for mediating cellular differentiation during development and allows adjustment to altered environmental conditions throughout life. Work over the last decade has begun to elucidate the way that extra- and intra-cellular signals lead to changes in gene expression patterns by directly modulating the function of chromatin-associated proteins. Here, we review key signaling-to-chromatin pathways that are specifically thought to target Polycomb and Trithorax group complexes, a classic example of epigenetically acting gene silencers and activators important in development, stem cell differentiation and cancer. We discuss the influence that signals triggered by kinase cascades, metabolic fluctuations and cell-cycle dynamics have on the function of these protein complexes. Further investigation into these pathways will be important for understanding the mechanisms that maintain epigenetic stability and those that promote epigenetic plasticity.
Collapse
Affiliation(s)
- Addie Kolybaba
- Ludwig Maximilians University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152, Planegg-Martinsried, Germany
| | | |
Collapse
|
286
|
Bustaffa E, Stoccoro A, Bianchi F, Migliore L. Genotoxic and epigenetic mechanisms in arsenic carcinogenicity. Arch Toxicol 2014; 88:1043-67. [PMID: 24691704 DOI: 10.1007/s00204-014-1233-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Arsenic is a human carcinogen with weak mutagenic properties that induces tumors through mechanisms not yet completely understood. People worldwide are exposed to arsenic-contaminated drinking water, and epidemiological studies showed a high percentage of lung, bladder, liver, and kidney cancer in these populations. Several mechanisms by which arsenical compounds induce tumorigenesis were proposed including genotoxic damage and chromosomal abnormalities. Over the past decade, a growing body of evidence indicated that epigenetic modifications have a role in arsenic-inducing adverse effects on human health. The main epigenetic mechanisms are DNA methylation in gene promoter regions that regulate gene expression, histone tail modifications that regulate the accessibility of transcriptional machinery to genes, and microRNA activity (noncoding RNA able to modulate mRNA translation). The "double capacity" of arsenic to induce mutations and epimutations could be the main cause of arsenic-induced carcinogenesis. The aim of this review is to better clarify the mechanisms of the initiation and/or the promotion of arsenic-induced carcinogenesis in order to understand the best way to perform an early diagnosis and a prompt prevention that is the key point for protecting arsenic-exposed population. Studies on arsenic-exposed population should be designed in order to examine more comprehensively the presence and consequences of these genetic/epigenetic alterations.
Collapse
Affiliation(s)
- Elisa Bustaffa
- Unit of Environmental Epidemiology and Diseases Registries, Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56123, Pisa, Italy
| | | | | | | |
Collapse
|
287
|
Alessandria L, Schilirò T, Degan R, Traversi D, Gilli G. Cytotoxic response in human lung epithelial cells and ion characteristics of urban-air particles from Torino, a northern Italian city. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:5554-64. [PMID: 24407785 DOI: 10.1007/s11356-013-2468-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 12/16/2013] [Indexed: 05/26/2023]
Abstract
Recently, much attention has been devoted to urban air pollution because epidemiological studies have reported health impacts related to particulate matter (PM). PM10 and PM2.5 were collected during different seasons in Torino, a northern Italian city, and were characterised by inorganic chemical species (secondary particulates and bio-available iron). The biological effects of aqueous and organic solvent PM extracts on human epithelial lung A549 were evaluated, and the effects on cell proliferation and lactate dehydrogenase (LDH) release were assayed. The average PM10 concentration during the sampling period was 47.9 ± 18.0 μg/m(3); the secondary particles accounted for 49 % ± 9 % of the PM10 total mass, and the bio-available iron concentration was 0.067 ± 0.045 μg/m(3). The PM2.5/PM10 ratio in Torino ranged from 0.47 to 0.90 and was higher in cold months than in warm months. The PM10 and PM2.5 extracts inhibited cell proliferation and induced LDH release in a dose-dependent manner with a seasonal trend. The PM10 extract had a stronger effect on LDH release, whereas the PM2.5 extract more strongly inhibited cell proliferation. No significant differences were observed in the effects induced by the two extracts, and no significant correlations were found between the biological effects and the PM components evaluated in this study, thus emphasising the importance of the entire mixture in inducing a cytotoxic response.
Collapse
Affiliation(s)
- Luca Alessandria
- Department of Public Health and Pediatrics, University of Torino, Piazza Polonia, 94, 10126, Torino, Italy
| | | | | | | | | |
Collapse
|
288
|
Qi Q, Bie P. Different roles of hepatic hypothermic ischemia and ischemic preconditioning in chemically induced hepatocarcinogenesis in rats. J Surg Res 2014; 189:213-21. [PMID: 24725680 DOI: 10.1016/j.jss.2014.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/28/2014] [Accepted: 03/05/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hepatic ischemia-reperfusion (IR) injury, an unfavorable complication of hepatectomy, could be prevented by hypothermic ischemia and ischemic preconditioning (IPC). However, the effects of these two approaches on hepatocarcinogenesis have not been examined. The aim of the study was to investigate roles of hypothermic ischemia and IPC in a chemically induced rat liver tumor model. METHODS Twenty-four Sprague-Dawley rats were treated with diethylnitrosamine and phenobarbital to induce hepatocellular carcinoma. Rats underwent hepatic ischemic injury, hypothermic ischemia, and IPC. Twenty-eight-wk-old rats were sacrificed to evaluate the morbidity and growth of liver tumor. Cytokines were measured at the protein and messenger RNA level. RESULTS IR injury significantly promoted liver tumor development. Intriguingly, hypothermic ischemia, but not IPC, delayed liver carcinogenesis, although both of them suppressed the hepatic IR injury. IPC-treated rats showed elevated interleukin (IL)-6 concentration in the serum and messenger RNA expression in liver. In addition, higher levels of IL-6 activated signal transducer and activator of transcription 3 in the liver of IPC-treated rats. The hepatic expression of target genes of signal transducer and activator of transcription 3 signaling, cyclin D1, c-myc, c-fos, and c-jun, all of which might participate in tumor progression, increased in IPC group, compared with that of IR group. CONCLUSIONS These data indicated hypothermic ischemia could ameliorate both IR injury and liver tumor development. However, IPC, another effective method to prevent hepatic IR injury, might exacerbate liver tumor growth. The elevated level of IL-6 was one of the reasons for the different effects of hypothermic ischemia and IPC on hepatocarcinogenesis in rats.
Collapse
Affiliation(s)
- Qingan Qi
- Southwest Hospital and Institute of Hepatobilitary Surgery, Third Military Medical University, Chongqing, China
| | - Ping Bie
- Southwest Hospital and Institute of Hepatobilitary Surgery, Third Military Medical University, Chongqing, China.
| |
Collapse
|
289
|
Callero MA, Luzzani GA, De Dios DO, Bradshaw TD, Perez AIL. Biomarkers of sensitivity to potent and selective antitumor 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F203) in ovarian cancer. J Cell Biochem 2014; 114:2392-404. [PMID: 23696052 DOI: 10.1002/jcb.24589] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/01/2013] [Indexed: 12/20/2022]
Abstract
2-(4-Amino-3-methylphenyl)-5-fluorobenzothiazole (5F203, NSC 703786) lysylamide belongs to a novel mechanistic class of antitumor agents. It elicits activity against ovarian, breast, kidney and colorectal cancer models. In sensitive breast cancer cells, 5F203 activates aryl hydrocarbon receptor (AhR) signaling. Herein, we evaluate the role of AhR in 5F203 activity in two ovarian cancer cell lines: IGROV-1 (sensitive to 5F203), SKOV-3 (resistant to this agent). In addition, cancer cells have been isolated from ascites fluid of ovarian cancer patients; sensitivity to 5F203 and concurrent AhR signal transduction has been examined in ascites-isolated ovarian cancer patients' cells. 5F203 induced enhanced CYP1A1 expression, AhR translocation and ROS formation in IGROV-1 cells and ascites-isolated ovarian cancer cells that were sensitive to 5F203. In IGROV-1 cells 5F203-induced ROS formation was accompanied by JNK, ERK and P38MAPK phosphorylation, DNA damage and cell cycle arrest prior to apoptosis. In contrast, 5F203 failed to induce CYP1A1 expression, AhR translocation or oxidative stress in 5F203-resistant SKOV-3 cells, or in ovarian cancer ascites cells inherently resistant to this agent. We propose that AhR may represent a new molecular target in the treatment of ovarian tumors and 5F203 may exemplify a potential novel treatment. Furthermore, putative biomarkers of sensitivity to this agent have been identified.
Collapse
Affiliation(s)
- Mariana A Callero
- National Scientific Council (CONICET), Ciudad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
290
|
Benzene-poly-carboxylic acid complex, a novel anti-cancer agent induces apoptosis in human breast cancer cells. PLoS One 2014; 9:e85156. [PMID: 24523856 PMCID: PMC3921106 DOI: 10.1371/journal.pone.0085156] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/23/2013] [Indexed: 01/03/2023] Open
Abstract
Some cases of breast cancer are composed of clones of hormonal-independent growing cells, which do not respond to therapy. In the present study, the effect of Benzene-Poly-Carboxylic Acid Complex (BP-C1) on growth of human breast-cancer cells was tested. BP-C1 is a novel anti-cancer complex of benzene-poly-carboxylic acids with a very low concentration of cis-diammineplatinum (II) dichloride. Human breast cancer cells, MCF-7 and T47D, were used. Cell viability was detected by XTT assay and apoptosis was detected by Flow Cytometry and by annexin V/FITC/PI assay. Caspases were detected by western blot analysis and gene expression was measured by using the Applied Biosystems® TaqMan® Array Plates. The results showed that exposure of the cells to BP-C1 for 48 h, significantly (P<0.001) reduced cell viability, induced apoptosis and activated caspase 8 and caspace 9. Moreover, gene expression experiments indicated that BP-C1 increased the expression of pro-apoptotic genes (CASP8AP1, TNFRSF21, NFkB2, FADD, BCL10 and CASP8) and lowered the level of mRNA transcripts of inhibitory apoptotic genes (BCL2L11, BCL2L2 and XIAP. These findings may lead to the development of new therapeutic strategies for treatment of human cancer using BP-C1 analog.
Collapse
|
291
|
Gordillo GM, Biswas A, Khanna S, Pan X, Sinha M, Roy S, Sen CK. Dicer knockdown inhibits endothelial cell tumor growth via microRNA 21a-3p targeting of Nox-4. J Biol Chem 2014; 289:9027-38. [PMID: 24497637 DOI: 10.1074/jbc.m113.519264] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miR) are emerging as biomarkers and potential therapeutic targets in tumor management. Endothelial cell tumors are the most common soft tissue tumors in infants, yet little is known about the significance of miR in regulating their growth. A validated mouse endothelial cell (EOMA) tumor model was used to demonstrate that post-transcriptional gene silencing of dicer, the enzyme that converts pre-miR to mature miR, can prevent tumor formation in vivo. Tumors were formed in eight of eight mice injected with EOMA cells transfected with control shRNA but formed in only four of ten mice injected with EOMA cells transfected with dicer shRNA. Tumors that formed in the dicer shRNA group were significantly smaller than tumors in the control group. This response to dicer knockdown was mediated by up-regulated miR 21a-3p activity targeting the nox-4 3'-UTR. EOMA cells were transfected with miR 21a-3p mimic and luciferase reporter plasmids containing either intact nox-4 3'-UTR or with mutation of the proposed 3'-UTR miR21a-3p binding sites. Mean luciferase activity was decreased by 85% in the intact compared with the site mutated vectors (p < 0.01). Attenuated Nox-4 activity resulted in decreased cellular hydrogen peroxide production and decreased production of oxidant-inducible monocyte chemoattractant protein-1, which we have previously shown to be critically required for endothelial cell tumor formation. These findings provide the first evidence establishing the significance of dicer and microRNA in promoting endothelial cell tumor growth in vivo.
Collapse
|
292
|
Ward A, Hudson JW. p53-Dependent and cell specific epigenetic regulation of the polo-like kinases under oxidative stress. PLoS One 2014; 9:e87918. [PMID: 24498222 PMCID: PMC3909268 DOI: 10.1371/journal.pone.0087918] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 01/01/2014] [Indexed: 12/27/2022] Open
Abstract
The polo-like kinase (PLKs) family, consisting of five known members, are key regulators of important cell cycle processes, which include mitotic entry, centrosome duplication, spindle assembly, and cytokinesis. The PLKs have been implicated in a variety of cancers, such as hepatocellular carcinoma (HCC), with PLK1 typically overexpressed and PLKs 2-5 often downregulated. Altered expression of the PLKs in malignancy is often correlated with aberrant promoter methylation. Epigenetic marks are dynamic and can be modified in response to external environmental stimuli. The aim of our study was to determine if oxidative stress, a common feature of solid tumours, would induce changes to the promoter methylation of the PLKs resulting in changes in expression. We examined the promoter methylation status via MSP and subsequent expression levels of the PLK family members under exposure to hypoxic conditions or reactive oxygen species (ROS). Interestingly, murine embryonic fibroblasts exposed to hypoxia and ROS displayed significant hypermethylation of Plk1 and Plk4 promoter regions post treatment. Corresponding proteins were also depleted by 40% after treatment. We also examined the HCC-derived cell lines HepG2 and Hep3B and found that for PLK1 and PLK4, the increase in hypermethylation was correlated with the presence of functional p53. In p53 wild-type cells, HepG2, both PLK1 and PLK4 were repressed with treatment, while in the p53 null cell line, Hep3B, PLK4 protein was elevated in the presence of hypoxia and ROS. This was also the case for ROS-treated, p53 null, osteosarcoma cells, Saos-2, where the PLK4 promoter became hypomethylated and protein levels were elevated. Our data supports a model in which the PLKs are susceptible to epigenetic changes induced by microenvironmental cues and these modifications may be p53-dependent. This has important implications in HCC and other cancers, where epigenetic alterations of the PLKs could contribute to tumourigenesis and disease progression.
Collapse
Affiliation(s)
- Alejandra Ward
- Department of Biology, University of Windsor, Windsor, Ontario, Canada
| | - John W. Hudson
- Department of Biology, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
293
|
Jakhar R, Paul S, Park YR, Han J, Kang SC. 3,5,7,3',4'-pentamethoxyflavone, a quercetin derivative protects DNA from oxidative challenges: potential mechanism of action. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 131:96-103. [PMID: 24514458 DOI: 10.1016/j.jphotobiol.2014.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/08/2013] [Accepted: 01/07/2014] [Indexed: 01/10/2023]
Abstract
DNA protection is one of the most important strategies in cancer therapy. Since quercetin and its derivatives are found to be potent antioxidant agents, they are able to scavenge radicals significantly. Therefore, we focused on the DNA protection activity of 3,5,7,3',4'-pentamethoxyflavone (PMF), a quercetin derivative isolated from Kaemperia parviflora. Although, PMF was found to be a very poor antioxidant compound, still it could remarkably protect DNA from oxidative damage. DNA binding assay showed that PMF bound to the minor groove of DNA, which suggests a possible mechanism for its DNA protective effects. Cellular toxicity assay on RAW 264.7 macrophages showed this compound is very safe for therapeutic applications.
Collapse
Affiliation(s)
- Rekha Jakhar
- Department of Biotechnology, Daegu University, Kyongsan, Kyongbok 712-714, Republic of Korea
| | - Souren Paul
- Department of Biotechnology, Daegu University, Kyongsan, Kyongbok 712-714, Republic of Korea
| | - Young Rong Park
- Metalloenzyme Research Group and School of Biological Sciences, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Jaehong Han
- Metalloenzyme Research Group and School of Biological Sciences, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Kyongsan, Kyongbok 712-714, Republic of Korea.
| |
Collapse
|
294
|
Soubry A, Hoyo C, Jirtle RL, Murphy SK. A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays 2014; 36:359-71. [PMID: 24431278 PMCID: PMC4047566 DOI: 10.1002/bies.201300113] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Literature on maternal exposures and the risk of epigenetic changes or diseases in the offspring is growing. Paternal contributions are often not considered. However, some animal and epidemiologic studies on various contaminants, nutrition, and lifestyle-related conditions suggest a paternal influence on the offspring's future health. The phenotypic outcomes may have been attributed to DNA damage or mutations, but increasing evidence shows that the inheritance of environmentally induced functional changes of the genome, and related disorders, are (also) driven by epigenetic components. In this essay we suggest the existence of epigenetic windows of susceptibility to environmental insults during sperm development. Changes in DNA methylation, histone modification, and non-coding RNAs are viable mechanistic candidates for a non-genetic transfer of paternal environmental information, from maturing germ cell to zygote. Inclusion of paternal factors in future research will ultimately improve the understanding of transgenerational epigenetic plasticity and health-related effects in future generations.
Collapse
Affiliation(s)
- Adelheid Soubry
- Epidemiology Research Group, Department of Public Health and Primary Care, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
295
|
Sung MK, Bae YJ. Iron, Oxidative Stress, and Cancer. Cancer 2014. [DOI: 10.1016/b978-0-12-405205-5.00013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
296
|
Neilson HK, Conroy SM, Friedenreich CM. The Influence of Energetic Factors on Biomarkers of Postmenopausal Breast Cancer Risk. Curr Nutr Rep 2013; 3:22-34. [PMID: 24563822 PMCID: PMC3921460 DOI: 10.1007/s13668-013-0069-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Strong and consistent evidence exists that physical activity reduces breast cancer risk by 10-25 %, and several proposed biologic mechanisms have now been investigated in randomized, controlled, exercise intervention trials. Leading hypothesized mechanisms relating to postmenopausal breast cancer include adiposity, endogenous sex hormones, insulin resistance, and chronic low-grade inflammation. In addition, other pathways are emerging as potentially important, including those involving oxidative stress and telomere length, global DNA hypomethylation, immune function, and vitamin D exposure. Recent exercise trials in overweight/obese postmenopausal women implicate weight loss as a mechanism whereby exercise induces favorable changes in circulating estradiol levels and other biomarkers as well. Still it is plausible that some exercise-induced biomarker changes do not require loss of body fat, whereas others depend on abdominal fat loss. We highlight the latest findings from randomized, controlled trials of healthy postmenopausal women, relating exercise to proposed biomarkers for postmenopausal breast cancer risk.
Collapse
Affiliation(s)
- Heather K Neilson
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada
| | - Shannon M Conroy
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada
| | - Christine M Friedenreich
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada ; Department of Community Health Sciences, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N2 Canada ; Department of Oncology, Faculty of Medicine, University of Calgary, 1331 29 St. N.W., Calgary, Alberta T2N 4N2 Canada
| |
Collapse
|
297
|
Samoylenko A, Hossain JA, Mennerich D, Kellokumpu S, Hiltunen JK, Kietzmann T. Nutritional countermeasures targeting reactive oxygen species in cancer: from mechanisms to biomarkers and clinical evidence. Antioxid Redox Signal 2013; 19:2157-96. [PMID: 23458328 PMCID: PMC3869543 DOI: 10.1089/ars.2012.4662] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 02/08/2013] [Accepted: 03/01/2013] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) exert various biological effects and contribute to signaling events during physiological and pathological processes. Enhanced levels of ROS are highly associated with different tumors, a Western lifestyle, and a nutritional regime. The supplementation of food with traditional antioxidants was shown to be protective against cancer in a number of studies both in vitro and in vivo. However, recent large-scale human trials in well-nourished populations did not confirm the beneficial role of antioxidants in cancer, whereas there is a well-established connection between longevity of several human populations and increased amount of antioxidants in their diets. Although our knowledge about ROS generators, ROS scavengers, and ROS signaling has improved, the knowledge about the direct link between nutrition, ROS levels, and cancer is limited. These limitations are partly due to lack of standardized reliable ROS measurement methods, easily usable biomarkers, knowledge of ROS action in cellular compartments, and individual genetic predispositions. The current review summarizes ROS formation due to nutrition with respect to macronutrients and antioxidant micronutrients in the context of cancer and discusses signaling mechanisms, used biomarkers, and its limitations along with large-scale human trials.
Collapse
Affiliation(s)
- Anatoly Samoylenko
- Department of Biochemistry, Biocenter Oulu, University of Oulu, Oulu, Finland
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Jubayer Al Hossain
- Department of Biochemistry, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Daniela Mennerich
- Department of Biochemistry, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sakari Kellokumpu
- Department of Biochemistry, Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Thomas Kietzmann
- Department of Biochemistry, Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
298
|
Expression and function of methylthioadenosine phosphorylase in chronic liver disease. PLoS One 2013; 8:e80703. [PMID: 24324622 PMCID: PMC3855635 DOI: 10.1371/journal.pone.0080703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 10/07/2013] [Indexed: 12/21/2022] Open
Abstract
To study expression and function of methylthioadenosine phosphorylase (MTAP), the rate-limiting enzyme in the methionine and adenine salvage pathway, in chronic liver disease.
Collapse
|
299
|
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer death, and its development is influenced by the status of inflammation and oxidative stress in the liver. Although oxidative stress might induce genetic changes and play a role in HCC development, many epigenetic alterations have also been reported in this type of tumor, suggesting the importance of epigenetic instability in hepatocarcinogenesis. Epigenetic instability results in 2 types of DNA alterations: hypermethylation of the promoter of tumor suppressor genes (TSGs), and hypomethylation of nonpromoter CpG, such as repetitive elements and satellite DNA. The former causes transcriptional inactivation of TSGs, while the latter reportedly induces chromosomal instability and an abnormal activation of oncogenes as well as mobile genetic elements. Oxidative stress could induce epigenetic instability and inactivate TSGs through the recruitment of the polycomb repressive complex to the promoter sequence carrying DNA damage induced by oxidation. Inflammatory cytokines from immune cells also reportedly induce expression of several histone and DNA modulators. On the other hand, DNA oxidation could lead to activation of DNA repair pathways and affect the binding of methyl cytosine-binding protein to DNA, which could cause DNA hypomethylation. The decrease of the level of methyl group donors also contributes to the alteration in the methylation status. These mechanisms should act in concert and induce epigenetic instability, leading to HCC.
Collapse
Affiliation(s)
- Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kinki University School of Medicine, Osakasayama, Japan
| | | |
Collapse
|
300
|
Aoi J, Endo M, Kadomatsu T, Miyata K, Ogata A, Horiguchi H, Odagiri H, Masuda T, Fukushima S, Jinnin M, Hirakawa S, Sawa T, Akaike T, Ihn H, Oike Y. Angiopoietin-like protein 2 accelerates carcinogenesis by activating chronic inflammation and oxidative stress. Mol Cancer Res 2013; 12:239-49. [PMID: 24258150 DOI: 10.1158/1541-7786.mcr-13-0336] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Chronic inflammation has received much attention as a risk factor for carcinogenesis. We recently reported that Angiopoietin-like protein 2 (Angptl2) facilitates inflammatory carcinogenesis and metastasis in a chemically induced squamous cell carcinoma (SCC) of the skin mouse model. In particular, we demonstrated that Angptl2-induced inflammation enhanced susceptibility of skin tissues to "preneoplastic change" and "malignant conversion" in SCC development; however, mechanisms underlying this activity remain unclear. Using this model, we now report that transgenic mice overexpressing Angptl2 in skin epithelial cells (K14-Angptl2 Tg mice) show enhanced oxidative stress in these tissues. Conversely, in the context of this model, Angptl2 knockout (KO) mice show significantly decreased oxidative stress in skin tissue as well as a lower incidence of SCC compared with wild-type mice. In the chemically induced SCC model, treatment of K14-Angptl2 Tg mice with the antioxidant N-acetyl cysteine (NAC) significantly reduced oxidative stress in skin tissue and the frequency of SCC development. Interestingly, K14-Angptl2 Tg mice in the model also showed significantly decreased expression of mRNA encoding the DNA mismatch repair enzyme Msh2 compared with wild-type mice and increased methylation of the Msh2 promoter in skin tissues. Msh2 expression in skin tissues of Tg mice was significantly increased by NAC treatment, as was Msh2 promoter demethylation. Overall, this study strongly suggests that the inflammatory mediator Angptl2 accelerates chemically induced carcinogenesis through increased oxidative stress and decreased Msh2 expression in skin tissue. IMPLICATIONS Angptl2-induced inflammation increases susceptibility to microenvironmental changes, allowing increased oxidative stress and decreased Msh2 expression; therefore, Angptl2 might be a target to develop new strategies to antagonize these activities in premalignant tissue.
Collapse
Affiliation(s)
- Jun Aoi
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|