1
|
Carrall JA, Lie W, Lambert JM, Harris HH, Lai B, Dillon CT. Optimizing Arsenic Therapy by Selectively Targeting Leukemia Cells. J Med Chem 2023; 66:12101-12114. [PMID: 37594965 DOI: 10.1021/acs.jmedchem.3c00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
Arsenic, in the simple form of arsenic trioxide, is currently marketed for the treatment of acute promyelocytic leukemia. Due to the multifaceted mechanisms of action of arsenic, it has also shown promise in other types of leukemias but is hindered by its toxic effects toward normal cells. This research has aimed to determine whether tumor-homing peptide complexes of arsenic can be designed and developed to strategically target specific cancers. The end goal is to achieve dose reduction and decreased side effects of the resultant arsenic therapeutic agent. In this article, we present the synthesis, characterization, and stability studies of a new class of As-peptide complexes designed to target leukemia. In vitro biological studies of the most stable complex show 1000 times greater toxicity toward leukemia cells over human blood cells, indicating potential for progression to in vivo studies.
Collapse
Affiliation(s)
- Judith A Carrall
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong 2522, New South Wales, Australia
| | - Wilford Lie
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong 2522, New South Wales, Australia
| | - Jacob M Lambert
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong 2522, New South Wales, Australia
- Molecular Horizons, University of Wollongong, Wollongong 2522, New South Wales, Australia
| | - Hugh H Harris
- School of Physical Sciences, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Barry Lai
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont 60439, Illinois, United States
| | - Carolyn T Dillon
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong 2522, New South Wales, Australia
- Molecular Horizons, University of Wollongong, Wollongong 2522, New South Wales, Australia
| |
Collapse
|
2
|
Mamgain R, Singh FV. Selenium-Based Fluorescence Probes for the Detection of Bioactive Molecules. ACS ORGANIC & INORGANIC AU 2022; 2:262-288. [PMID: 36855593 PMCID: PMC9954296 DOI: 10.1021/acsorginorgau.1c00047] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chemistry of organoselenium reagents have now become an important tool of synthetic organic and medicinal chemistry. These reagents activate the olefinic double bonds and used to archive the number of organic transformations under mild reaction conditions. A number of organoselenium compounds have been identified as potent oxidants. Recently, various organoselenium species have been employed as chemical sensors for detecting toxic metals. Moreover, a number of selenium-based fluorescent probes have been developed for detecting harmful peroxides and ROS. In this review article, the synthesis of selenium-based fluorescent probes will be covered including their application in the detection of toxic metals and harmful peroxides including ROS.
Collapse
Affiliation(s)
- Ritu Mamgain
- Chemistry
Division, School of Advanced Sciences (SAS),
Vellore Institute of Technology-Chennai, Vandalur-Kelambakkam Road, Chennai 600127, Tamil
Nadu, India
| | - Fateh V. Singh
- Chemistry
Division, School of Advanced Sciences (SAS),
Vellore Institute of Technology-Chennai, Vandalur-Kelambakkam Road, Chennai 600127, Tamil
Nadu, India,
| |
Collapse
|
3
|
Yen CM, Tsai CW, Chang WS, Yang YC, Hung YW, Lee HT, Shen CC, Sheu ML, Wang JY, Gong CL, Cheng WY, Bau DAT. Novel Combination of Arsenic Trioxide (As 2O 3) Plus Resveratrol in Inducing Programmed Cell Death of Human Neuroblastoma SK-N-SH Cells. Cancer Genomics Proteomics 2018; 15:453-460. [PMID: 30343279 DOI: 10.21873/cgp.20104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/13/2018] [Accepted: 09/19/2018] [Indexed: 12/23/2022] Open
Abstract
AIM Arsenic trioxide (As2O3), known as pi-shuang and the most toxic compound in traditional Chinese medicine, has been used as an antitumor agent for thousands of years. Resveratrol (3,5,4'-trihydroxy-trans-stilbene) is a natural phenol that has significant anti-bacterial, anti-fungaI and antiaging activities. Our study aimed to examine the combined anticancer effects of As2O3 and resveratrol against human neuroblastoma SK-N-SH cells, and elucidate the underlying intracellular signaling. MATERIALS AND METHODS SK-N-SH cells were treated with an extremely low-dose (2-4 μM) of As2O3 alone or combined with 75 μg/ml resveratrol for further comparisons. Cell viability, apoptotic signaling as well as synergistic cytotoxic effects were estimated using the MTT assay, microscopy observation, flow cytometric analysis for loss of mitochondrial membrane potential (MMP) and reactive oxygen species (ROS), and typical quantitative western blotting analysis. Student's t-test, and one- and two-way analysis of variance (ANOVA) were used for examination of significant differences. RESULTS The combined treatment was more effective than single treatment of As2O3 or resveratrol alone in suppressing cell viability, which correlated with the elevation of ROS levels. The intracellular mechanisms of cytotoxicity of As2O3 plus resveratrol were revealed as ROS accumulation and relative decrease of MMP, leading to activation of caspase-3 and -9, but not of caspase-1, -7 and-8. Combination treatment reduced the expression of B-cell lymphoma 2 (BCL2), BH3 interacting domain death agonist (BID), and BCL-x/L. CONCLUSION Combined treatment at extremely low concentration of two agents from natural products, As2O3 and resveratrol, has high potential as a cocktail of anticancer drugs for neuroblastoma.
Collapse
Affiliation(s)
- Chun-Ming Yen
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, R.O.C.,Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Chin Yang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Wen Hung
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Hsu-Tung Lee
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Chiung-Chyi Shen
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, R.O.C
| | - Ju-Yu Wang
- Basic Medical Science, Department of Nursing, Hung Kuang University, Taichung, Taiwan, R.O.C
| | - Chi-Li Gong
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C.
| | - Wen-Yu Cheng
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C. .,Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
4
|
Szekeres LI, Gyurcsik B, Kiss T, Kele Z, Jancsó A. Interaction of Arsenous Acid with the Dithiol-Type Chelator British Anti-Lewisite (BAL): Structure and Stability of Species Formed in an Unexpectedly Complex System. Inorg Chem 2018; 57:7191-7200. [PMID: 29856616 DOI: 10.1021/acs.inorgchem.8b00894] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
British anti-Lewisite (2,3-dimerkaptopropan-1-ol, dimercaprol, BAL) is one of the best-known chelator-type therapeutic agents against toxic metal ions and metalloids, especially arsenicals. Surprisingly, the mechanisms of action at the molecular level, as well as the coordination features of this traditional drug toward various arsenicals, are still poorly revealed. The present study on the interaction of arsenous acid (H3AsO3) with BAL, involving UV and NMR titrations, electrospray ionization mass spectrometry, and 2D NMR experiments combined with MP2 calculations, demonstrates that the reaction of H3AsO3 with BAL at pH = 7.0 results in a more complex speciation than was assumed before. The three reactive hydroxyl groups of H3AsO3 allow for interaction with three thiol moieties via condensation reaction, leading to the observed AsBAL2 and As2BAL3 complexes besides the AsBAL species. This indicates the strong propensity of inorganic As(III) to saturate its coordination sphere with thiolate groups. The alcoholic hydroxyl group of the ligand may also directly bind to As(III) in AsBAL. Compared to dithiothreitol or dithioeritritol, the preference of BAL to form complexes with such a tridentate binding mode is much lower owing to the more strained bridged bicyclic structure with an αAsSC < 90° bond angle and an unfavorable condensed boat-type six-membered ring. On the basis of the NMR data, the predominating, bidentately bound AsBAL species, including a five-membered chelate ring, exists in rapidly interconverting envelope forms of E and Z stereoisomers. The conditional stability constants calculated for the three macrospecies from a series of UV data [log βpH=7.0 = 6.95 (AsBAL), 11.56 (AsBAL2), and 22.73 (As2BAL3)] reflect that BAL is still the most efficient, known, dithiol-type chelator of H3AsO3.
Collapse
Affiliation(s)
- Levente I Szekeres
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7 , Szeged H-6720 , Hungary
| | - Béla Gyurcsik
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7 , Szeged H-6720 , Hungary
| | - Tamás Kiss
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7 , Szeged H-6720 , Hungary
| | - Zoltán Kele
- Department of Medical Chemistry , University of Szeged , Dóm tér 8 , Szeged H-6720 , Hungary
| | - Attila Jancsó
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7 , Szeged H-6720 , Hungary
| |
Collapse
|
5
|
Arslan-Acaroz D, Zemheri F, Demirel HH, Kucukkurt I, Ince S, Eryavuz A. In vivo assessment of polydatin, a natural polyphenol compound, on arsenic-induced free radical overproduction, gene expression, and genotoxicity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:2614-2622. [PMID: 29130132 DOI: 10.1007/s11356-017-0391-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 10/02/2017] [Indexed: 06/07/2023]
Abstract
Arsenic (As) is a well-known contaminant of global groundwater. Its exposure causes several hazardous effects on animals and human via oxidative stress. The present study examined the effect of polydatin (PD) on free radical overproduction in rats exposed to As. Thirty-five male rats randomly allocated into five equal groups. To the control group, physiological saline was given orally and to the second group only 100 mg/L As was given by drinking water for 60 days. The other groups were treated with As (100 mg/L) and PD orally at 50, 100, and 200 mg/kg/day, respectively. Treatment with As enhanced malondialdehyde level but decreased glutathione level in blood, liver, kidney, brain, lung, and heart of rats. Also, As decreased superoxide dismutase and catalase activities of erythrocyte, liver, kidney, brain, lung, and heart in rats. Furthermore, As treatment gave rise to increased DNA damage and gene expressions of interleukin 1 beta (IL-1β), nuclear factor kappa beta (NFκB), p53, and tumor necrosis factor-α (TNF-α) in the lung, brain, kidney, and liver. However, treatment of PD ameliorated As-exposed lipid peroxidation, antioxidant enzymes activities, DNA damage, gene expressions, and histopathological changes in tissues. In conclusion, PD has a dose-dependent protective effect on lipid peroxidation and antioxidant defense mechanism in rats against As exposure.
Collapse
Affiliation(s)
- Damla Arslan-Acaroz
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| | - Fahriye Zemheri
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Bartin University, 74100, Bartin, Turkey
| | | | - Ismail Kucukkurt
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| | - Sinan Ince
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey.
| | - Abdullah Eryavuz
- Department of Physiology, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| |
Collapse
|
6
|
Tsai CW, Yang MD, Hsia TC, Chang WS, Hsu CM, Hsieh YH, Chung JG, Bau DT. Dithiothreitol enhanced arsenic-trioxide-induced cell apoptosis in cultured oral cancer cells via mitochondrial dysfunction and endoplasmic reticulum stress. ENVIRONMENTAL TOXICOLOGY 2017; 32:17-27. [PMID: 26494474 DOI: 10.1002/tox.22208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/02/2015] [Accepted: 10/04/2015] [Indexed: 06/05/2023]
Abstract
Arsenic is naturally occurring toxic metalloid and drinking As2 O3 containing water are recognized to be related to increased risk of neurotoxicity, liver injury, blackfoot disease, hypertension, and cancer. On the contrary, As2 O3 has been an ancient drug used in traditional Chinese medicine with substantial anticancer activities, especially in the treatment of acute promyelocytic leukemia as well as chronic wound healing. However, the cytotoxicity and detail mechanisms of As2 O3 action in solid cancer cells, such as oral cancer cells, are largely unknown. In this study, we have primarily cultured four pairs of tumor and nontumor cells from the oral cancer patients and treated the cells with As2 O3 alone or combined with dithiothreitol (DTT). The results showed that 0.5 μM As2 O3 plus 20 μM DTT caused a significant cell death of oral cancer cells but not the nontumor cells. Also As2 O3 plus DTT upregulated Bax and Bak, downregulated Bcl-2 and p53, caused a loss of mitochondria membrane potential in oral cancer cells. On the other way, As2 O3 also triggered endoplasmic reticulum stress and increased the levels of glucose-regulated protein 78, calpain 1 and 2. Our results suggest that DTT could synergistically enhance the effects of As2 O3 on killing oral cancer cells while nontoxic to the nontumor cells. The combination is promising for clinical practice in oral cancer therapy and worth further investigations. © 2015 Wiley Periodicals, Inc. Environ Toxicol 32: 17-27, 2017.
Collapse
Affiliation(s)
- Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Due Yang
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
- Department of Clinical Nutrition, China Medical University Hospital, Taichung, Taiwan
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
- Department of Intensive Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Chin-Mu Hsu
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institution of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
7
|
Kumazaki M, Ando H, Kakei M, Ushijima K, Taniguchi Y, Yoshida M, Yamato S, Washino S, Koshimizu TA, Fujimura A. α-Lipoic acid protects against arsenic trioxide-induced acute QT prolongation in anesthetized guinea pigs. Eur J Pharmacol 2013; 705:1-10. [PMID: 23474023 DOI: 10.1016/j.ejphar.2013.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 01/28/2013] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
Clinical use of arsenic trioxide (As₂O₃), which can induce the remission of relapsed or refractory acute promyelocytic leukemia, is often limited because of its cardiotoxicity. Symptoms of cardiotoxicity include acute cardiac conduction disturbances, such as QT prolongation. The present study was undertaken to evaluate the effects of α-lipoic acid (LA) on acute As₂O₃-induced ECG abnormalities (QTc interval prolongation) in anesthetized guinea pigs. Intravenous injection of As₂O₃ in guinea pigs caused QTc interval prolongation, which was significantly attenuated by co-treatment with LA (0.35, 3.5 and 35 mg/kg) in a dose-dependent manner. In isolated guinea pig cardiomyocytes, the decrease in IKs current induced by As₂O3 (1 μM) was rapidly restored to the basal level by the addition of LA (10 μM). Consistent with this finding, the As₂O₃-induced QTc interval prolongation was also improved rapidly by post-treatment with LA in guinea pigs. Electrospray ionization time-of-flight mass spectrometry analysis detected an expected peak of arsenic-LA complex in vitro, indicating that LA and As₂O3 form a new compound in vivo. In addition, pre-treatment with a chelating agent, British anti-Lewisite (BAL, 3.5 or 35 mg/kg), also attenuated the As₂O₃-induced QTc interval prolongation. In this study, co- and post-treatments with LA and pre-treatment with BAL ameliorated As₂O₃-induced acute QT prolongation in anesthetized guinea pigs. Because LA and probably BAL may bind to As₂O₃, these agents may exert protective effects through their chelating activity. Further studies are needed to determine whether LA is beneficial as a prophylactic or rescue agent for acute promyelocytic leukemia patients treated with As₂O₃.
Collapse
Affiliation(s)
- Masafumi Kumazaki
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Sharifi AM, Mousavi SH. Studying the effects of lead on DNA fragmentation and proapoptotic bax and antiapoptotic bcl-2 protein expression in PC12 cells. Toxicol Mech Methods 2012; 18:75-9. [PMID: 20020893 DOI: 10.1080/15376510701665814] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
ABSTRACT The nervous system is one of the most important targets of lead poisoning. Despite decades of study, the exact mechanism of lead toxicity has not been fully elucidated. One of the suggested mechanisms of lead toxicity is induction of apoptosis, which has not been shown yet in some neuronal cells such as pheochromocytoma cells (PC12). Therefore, the present study sought to examine the effect of lead poisoning on apoptosis in PC12 cells as a suitable model of neuronal cell study. The present results showed that lead could induce toxicity in PC12 cells after 24 hours with as little as 1 muM in a concentration-dependent manner. In Western blot analysis, the ratio of Bax/Bcl-2 protein expression in cells incubated with 3, 30, and 90 muM lead acetate significantly increased compared to controls. Additionally, a DNA laddering pattern in lead-treated cells was shown, which could indicate nuclear fragmentation. It might be concluded that lead could cause PC12 cell death, in which apoptosis or programmed cell death plays an important role.
Collapse
Affiliation(s)
- Ali M Sharifi
- Department of Pharmacology and Cellular and Molecular Research Center, School of Medicine, Iran University of Medical Sciences, P.O. Box 14155-6183, Tehran, Iran
| | | |
Collapse
|
9
|
Jia Q, Ha X, Yang Z, Hui L, Yang X. Oxidative stress: a possible mechanism for lead-induced apoptosis and nephrotoxicity. Toxicol Mech Methods 2012; 22:705-10. [PMID: 22894711 DOI: 10.3109/15376516.2012.718811] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lead-induced nephrotoxicity is a human health hazard problem. In this study, Human mesangial cells (HMCs) were treated with different concentration of lead acetate (5, 10, 20 μmol/l) in order to investigate the oxidative stress and apoptotic changes. It was revealed that lead acetate could induce a progressive loss in HMCs viability together with a significant increase in the number of apoptotic cells using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium (MTT) assay and flow cytometry, respectively. The apoptotic morphological changes induced by lead exposure in HMCs were demonstrated by PI-Hochest33342 staining. A DNA laddering pattern in lead-treated cells was shown, which could indicate nuclear fragmentation. In addition, lead acetate significantly increased the levels of malondialehyde (MDA) content and lactate dehydrogenase (LDH) activity. Therefore, it might be concluded that lead could promote HMCs' oxidative stress and apoptosis, which may be the chief mechanisms of lead-induced nephrotoxicity.
Collapse
Affiliation(s)
- Qinghua Jia
- Experimental Center of Medicine, Lanzhou General Hospital of Lanzhou Military, People's Liberation Army, Key Laboratory of Stem Cells and Gene Drug of Gansu Province, 333 Southern Binhe Road, Lanzhou 730050, China.
| | | | | | | | | |
Collapse
|
10
|
Kim SC, Park SJ, Lee JR, Seo JC, Yang CH, Byun SH. Cytoprotective Activity of Glycyrrhizae radix Extract Against Arsenite-induced Cytotoxicity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 5:165-71. [PMID: 18604262 PMCID: PMC2396482 DOI: 10.1093/ecam/nem014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Accepted: 01/16/2007] [Indexed: 11/14/2022]
Abstract
Licorice, Glycyrrhizae radix, is one of the herbal medicines in East Asia that has been commonly used for treating various diseases, including stomach disorders. This study investigated the effect of licorice on arsenite (As)-induced cytotoxicity in H4IIE cells, a rat hepatocyte-derived cell line. Cell viability was significantly diminished in As-treated H4IIE cells in a time and concentration-dependent manner. Furthermore, results from flow cytometric assay and DNA laddering in H4IIE cells showed that As treatment induced apoptotic cell death by activating caspase-3. Licorice (0.1 and 1.0 mg ml(-1)) treatment significantly inhibited cell death and the activity of caspase-3 in response to As exposure. These results demonstrate that licorice induced a cytoprotective effect against As-induced cell death by inhibition of caspase-3.
Collapse
Affiliation(s)
- Sang Chan Kim
- College of Oriental Medicine and Research Center for Biomedical Resources of Oriental Medicine, Daegu Haany University, 165 Sang-dong, Suseong-gu, Daegu 706-060, Korea
| | | | | | | | | | | |
Collapse
|
11
|
Watanabe T, Hirano S. Metabolism of arsenic and its toxicological relevance. Arch Toxicol 2012; 87:969-79. [PMID: 22811022 DOI: 10.1007/s00204-012-0904-5] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
Abstract
Arsenic is a worldwide environmental pollutant and a human carcinogen. It is well recognized that the toxicity of arsenicals largely depends on the oxidoreduction states (trivalent or pentavalent) and methylation levels (monomethyl, dimethyl, and trimethyl) that are present during the process of metabolism in mammals. However, presently, the specifics of the metabolic pathway of inorganic arsenicals have yet to be confirmed. In mammals, there are two possible mechanisms that have been proposed for the metabolic pathway of inorganic arsenicals, oxidative methylation, and glutathione conjugation. Oxidative methylation, which was originally proposed in fungi, is based on findings that arsenite (iAs(III)) is sequentially converted to monomethylarsonic acid (MMA(V)) and dimethylarsinic acid (DMA(V)) in both humans and in laboratory animals such as mice and rats. However, recent in vitro observations have demonstrated that arsenic is only methylated in the presence of glutathione (GSH) or other thiol compounds, which strongly suggests that arsenic is methylated in trivalent forms. The glutathione conjugation mechanism is supported by findings that have shown that most intracellular arsenicals are trivalent and excreted from cells as GSH conjugates. Since non-conjugated trivalent arsenicals are highly reactive with thiol compounds and are easily converted to less toxic corresponding pentavalent arsenicals, the arsenic-glutathione conjugate stability may be the most important factor for determining the toxicity of arsenicals. In addition, "being a non-anionic form" also appears to be a determinant of the toxicity of oxo-arsenicals or thioarsenicals. The present review discusses both the metabolism of arsenic and the toxicity of arsenic metabolites.
Collapse
Affiliation(s)
- Takayuki Watanabe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-0856, Japan
| | | |
Collapse
|
12
|
Saad SY, Alkharfy KM, Arafah MM. Cardiotoxic effects of arsenic trioxide/imatinib mesilate combination in rats. J Pharm Pharmacol 2010; 58:567-73. [PMID: 16597375 DOI: 10.1211/jpp.58.4.0017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Cardiotoxicity is an important consideration in the evaluation of cancer chemotherapy, because chemotherapy-induced myocardial damage might be irreversible and lethal. This in-vivo study investigated the cardiotoxicity of either arsenic trioxide or imatinib mesilate, or a combination of both drugs, following repeated administration in male Wistar rats. Both arsenic trioxide and imatinib mesilate were administered daily at a dose of 5 mg kg−1 intraperitoneally and 30 mg kg−1 orally for 10 days, respectively. Cardiotoxicity was evaluated by biochemical and histopathological examination 48 h after the last dose. Treatment with either arsenic or imatinib, or both, resulted in significant increases in serum creatine kinase isoenzyme (CK-MB), glutathione peroxidase (GPx), lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) activity levels. Cardiac tissue of rats treated with arsenic showed significant increases in levels of reduced glutathione (GSH) content, GPx activity, malondialdehyde (MDA) and total nitrate/nitrite (NOx), whereas imatinib treatment significantly increased cardiac GSH content and MDA production level and decreased GPx activity level and NOx content. A combination of arsenic and imatinib produced significant increases in cardiac GSH content, GPx activity and MDA production levels, in addition to a reduction in NOx content. Combination arsenic/imatinib treatment extensively increased GPx activity and MDA production levels compared with imatinib treatment alone. Moreover, rats treated with arsenic or imatinib, or both, showed a significant increase in serum bilirubin, creatinine and urea levels. Histopathological examination of cardiac tissue of the combination-treated group revealed fibroblastic proliferation, myocardial disorganization and myocardial necrosis. Liver peroxidative alterations revealed that treatment with either arsenic or imatinib, or the two combined, increased levels of reduced-GSH and MDA production levels. However, imatinib treatment depleted liver GPx activity level contrary to treatment with the combination. Rats treated with arsenic alone or arsenic/imatinib combination showed significant elevation in liver NOx. In conclusion, both arsenic trioxide and imatinib mesilate might have significant cardiotoxicity and cardiac function should be monitored during treatment with them alone or in combination, as well as in the presence of pre-existing cardiac dysfunction.
Collapse
Affiliation(s)
- Sherif Y Saad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | | | | |
Collapse
|
13
|
Zhu B, Zhang X, Jia H, Li Y, Liu H, Tan W. A highly selective ratiometric fluorescent probe for 1,4-dithiothreitol (DTT) detection. Org Biomol Chem 2010; 8:1650-4. [DOI: 10.1039/b923754b] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Li H, Zhu X, Zhang Y, Xiang J, Chen H. Arsenic trioxide exerts synergistic effects with cisplatin on non-small cell lung cancer cells via apoptosis induction. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2009; 28:110. [PMID: 19664237 PMCID: PMC3225875 DOI: 10.1186/1756-9966-28-110] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Accepted: 08/08/2009] [Indexed: 11/10/2022]
Abstract
Background Despite multidisciplinary treatment, lung cancer remains a highly lethal disease due to poor response to chemotherapy. The identification of therapeutic agents with synergistic effects with traditional drugs is an alternative for lung cancer therapy. In this study, the synergistic effects of arsenic trioxide (As2O3) with cisplatin (DDP) on A549 and H460 non-small cell lung cancer (NSCLC) cells were explored. Methods A549 and H460 human lung cancer cells were treated with As2O3 and/or DDP. Cell growth curves, cell proliferation, cell cycle, and apoptosis of human cancer cell lines were determined by the 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) method, clonogenic assay, and flow cytometry (FCM). Apoptosis was further assessed by TUNEL staining. Cell cycle and apoptosis related protein p21, cyclin D1, Bcl-2, bax, clusterin, and caspase-3 were detected by western blot. Results MTT and clonogenic assay showed As2O3 within 10-2 μM to 10 μM exerted inhibition on the proliferation of NSCLC cells, and 2.5 μM As2O3 exerted synergistic inhibition on proliferation with 3 μg/ml DDP. The combination indices (CI) for A549 and H460 were 0.5 and 0.6, respectively, as confirmed by the synergism of As2O3 with DDP. FCM showed As2O3 did not affect the cell cycle. The G0/G1 fraction ranged from 57% to 62% for controlled A549 cells and cells treated with As2O3 and/or DDP. The G0/G1 fraction ranged from 37% to 42% for controlled H460 cells and cells treated with As2O3 and/or DDP. FCM and TUNEL staining illustrated that the combination of As2O3 and DDP provoked synergistic effects on apoptosis induction based on the analysis of the apoptosis index. Western blotting revealed that the expression of cell cycle related protein p21 and cyclin D1 were not affected by the treatments, whereas apoptosis related protein bax, Bcl-2, and clusterin were significantly regulated by As2O3 and/or DDP treatments compared with controls. The expression of caspase-3 in cells treated with the combination of As2O3 and DDP did not differ from that in cells treated with a single agent. Conclusion As2O3 exerted synergistic effects with DDP on NSCLC cells, and the synergistic effects were partly due to the induction of caspase-independent apoptosis.
Collapse
Affiliation(s)
- Hecheng Li
- Department of Thoracic Surgery, Fudan University Cancer Hospital/Cancer Institute, Shanghai, PR China.
| | | | | | | | | |
Collapse
|
15
|
Sharifi AM, Eslami H, Larijani B, Davoodi J. Involvement of caspase-8, -9, and -3 in high glucose-induced apoptosis in PC12 cells. Neurosci Lett 2009; 459:47-51. [DOI: 10.1016/j.neulet.2009.03.100] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 03/17/2009] [Accepted: 03/30/2009] [Indexed: 01/06/2023]
|
16
|
Solovieva ME, Solovyev VV, Kudryavtsev AA, Trizna YA, Akatov VS. Vitamin B12b enhances the cytotoxicity of dithiothreitol. Free Radic Biol Med 2008; 44:1846-56. [PMID: 18342018 DOI: 10.1016/j.freeradbiomed.2008.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 02/02/2008] [Accepted: 02/08/2008] [Indexed: 10/22/2022]
Abstract
It has been found previously that vitamin B12b amplifies significantly the cytotoxic effects of ascorbic acid by catalyzing the formation of reactive oxygen species, and the antioxidant dithiothreitol (DTT), in contrast to catalase, does not prevent the cytotoxicity. Therefore, in this study we examined whether B12b is able to enhance the cytotoxicity of DTT. It was revealed that B12b strongly increases the cytotoxic effect of DTT. Vitamin B12b added to DTT catalyzed the generation and drastic accumulation of hydrogen peroxide in culture medium to a concentration of 260 microM within 7 min. The extracellular oxidative burst induced by the combination of B12b and DTT (DTT + B12b) was accompanied by intracellular oxidative stress, the destabilization of lysosomes, and damage to DNA. The accumulation of DNA lesions led to the initiation of apoptotic cell death, including the activation of caspase-3 and the release of cytochrome c. The antioxidants pyruvate and catalase completely prevented the DTT + B12b-induced oxidative stress and cell death. The iron chelators desferrioxamine and phenanthroline prevented the geno- and cytotoxic action of the combination although they did not reduce the exogenous oxidative burst, indicating a key role for intracellular iron in the cytotoxicity of the combination. Thus, vitamin B12b dramatically enhances the cytotoxicity of DTT, catalyzing the generation of hydrogen peroxide and inducing extra- and intracellular oxidative stress, early destabilization of lysosomes, and iron-dependent DNA damage.
Collapse
Affiliation(s)
- Marina E Solovieva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | | | | | | | | |
Collapse
|
17
|
Kolozsi A, Lakatos A, Galbács G, Madsen AØ, Larsen E, Gyurcsik B. A pH-Metric, UV, NMR, and X-ray Crystallographic Study on Arsenous Acid Reacting with Dithioerythritol. Inorg Chem 2008; 47:3832-40. [DOI: 10.1021/ic7024439] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- András Kolozsi
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Andrea Lakatos
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Gábor Galbács
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Anders Østergaard Madsen
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Erik Larsen
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Béla Gyurcsik
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dóm tér 7, H-6701 Szeged, P.O. Box 440, Hungary, Biocoordinaton Chemistry Research Group of the Hungarian Academy of Sciences, University of Szeged, H-6701 Szeged, PO Box 440, Hungary, Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark, and Department of Natural Sciences, IGV, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| |
Collapse
|
18
|
Paul MK, Kumar R, Mukhopadhyay AK. Dithiothreitol abrogates the effect of arsenic trioxide on normal rat liver mitochondria and human hepatocellular carcinoma cells. Toxicol Appl Pharmacol 2007; 226:140-52. [PMID: 18022205 DOI: 10.1016/j.taap.2007.09.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 09/11/2007] [Accepted: 09/12/2007] [Indexed: 12/17/2022]
Abstract
Arsenic trioxide (ATO) is a known environmental toxicant and a potent chemotherapeutic agent. Significant correlation has been reported between consumption of arsenic-contaminated water and occurrence of liver cancer; moreover, ATO-treated leukemia patients also suffers from liver toxicity. Hence, modulation of ATO action may help to prevent populations suffering from arsenic toxicity as well as help reduce the drug-related side effects. Dithiothreitol (DTT) is a well-known dithiol agent reported to modulate the action of ATO. Controversial reports exist regarding the effect of DTT on ATO-induced apoptosis in leukemia cells. To the best of our knowledge, no report illustrates the modulatory effect of DTT on ATO-induced liver toxicity, the prime target for arsenic. Mitochondria serve as the doorway to apoptosis and have been implicated in ATO-induced cell death. Hence, we attempted to study the modulatory effect of DTT on ATO-induced dysfunction of mammalian liver mitochondria and human hepatocellular carcinoma cell line (Hep3B). We, for the first time, report that ATO produces complex I-mediated electron transfer inhibition, reactive oxygen species (ROS) generation, respiration inhibition, and ATO-induced ROS-mediated mitochondrial permeability transition (MPT) opening. DTT at low concentration (100 muM and less) prevents the effect of ATO-induced complex I-malfunctions. DTT protects mitochondria from ATO-mediated opening of MPT and membrane potential depolarization. DTT also prevented ATO-induced Hep3B cell death. Thus, at low concentrations DTT abrogates the effect of ATO on rat liver mitochondria and Hep3B cell line. Therefore, the present result suggests, that use of low concentration of dithiols as food supplement may prevent arsenic toxicity in affected population.
Collapse
Affiliation(s)
- Manash K Paul
- Department of Biology, Indian Institute of Science Education and Research, Mohali, MGSIPA Complex, Adjacent Sacred Heart School, Sector-26, Chandigarh-160019 [corrected] India.
| | | | | |
Collapse
|
19
|
Sharifi AM, Mousavi SH, Farhadi M, Larijani B. Study of High Glucose-Induced Apoptosis in PC12 Cells: Role of Bax Protein. J Pharmacol Sci 2007; 104:258-62. [PMID: 17652910 DOI: 10.1254/jphs.fp0070258] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Hyperglycemia, which occurs under the diabetic condition, induces serious diabetic complications. Diabetic neuropathies, affecting the autonomic, sensory, and motor peripheral nervous system, are among the most frequent complications of diabetes. Little is known about the direct toxic effect of high glucose concentrations on neuronal cells. Therefore in the present study, glucose-induced toxicity was studied in PC12 cells as an in vitro cellular model for diabetic neuropathy using the MTT assay. The possible role of apoptosis was also investigated in this toxicity. The result showed that a 3-fold increase in optimum glucose concentration for PC12 cells (13.5 mg/ml) significantly reduced cell viability after 48 h. In Western blot analysis, the ratio of Bax/Bcl-2 protein expression in cells treated with high glucose was significantly increased compared to controls. Additionally high glucose could induce a DNA ladder pattern in PC12 cells, a hallmark of apoptosis indicating nuclear fragmentation. From our present results, it may be concluded that high glucose can cause PC12 cell death, in which apoptosis plays an important role possibly by the mitochondrial pathway through higher expression of Bax pro-apoptotic protein.
Collapse
Affiliation(s)
- Ali M Sharifi
- Department of Pharmacology and Cellular and Molecular Research Center, School of Medicine, Iran University of Medical Sciences, P.O. Box 14155-6183, Tehran, Iran.
| | | | | | | |
Collapse
|
20
|
Jan KY, Wang TC, Ramanathan B, Gurr JR. Dithiol Compounds at Low Concentrations Increase Arsenite Toxicity. Toxicol Sci 2006; 90:432-9. [PMID: 16421177 DOI: 10.1093/toxsci/kfj108] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Inorganic trivalent arsenicals are vicinal thiol-reacting agents, and dithiothreitol (DTT) is a well-known dithiol agent. Interestingly, both decreasing and increasing effects of DTT on arsenic trioxide-induced apoptosis have been reported. We now provide data to show that, at high concentrations, DTT, dimercaptosuccinic acid (DMSA), and dimercaptopropanesulfonic acid (DMPS) decreased arsenic trioxide-induced apoptosis in NB4 cells, a human promyelocytic leukemia cell line. In contrast, at low concentrations DTT, DMSA, and DMPS increased the arsenic trioxide-induced apoptosis. DTT at a high concentration (3 mM) decreased, whereas at a low concentration (0.1 mM), it increased the cell growth inhibition of arsenic trioxide, methylarsonous acid (MMA(III)), and dimethylarsinous acid (DMA(III)) in NB4 cells. DMSA and DMPS are currently used as antidotes for acute arsenic poisoning. These two dithiol compounds also show an inverse-hormetic effect on arsenic toxicity in terms of DNA damage, micronucleus induction, apoptosis, and colony formation in experiments using human epithelial cell lines derived from arsenic target tissues such as the kidney and bladder. With the oral administration of dithiols, the concentrations of these dithiol compounds in the human body are likely to be low. Therefore, the present results suggest the necessity of reevaluating the therapeutic effect of these dithiol compounds for arsenic poisoning.
Collapse
Affiliation(s)
- Kun-Yan Jan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan, ROC
| | | | | | | |
Collapse
|
21
|
Blakytny R, Erkell LJ, Brunner G. Inactivation of active and latent transforming growth factor beta by free thiols: Potential redox regulation of biological action. Int J Biochem Cell Biol 2006; 38:1363-73. [PMID: 16531095 DOI: 10.1016/j.biocel.2006.01.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Revised: 01/26/2006] [Accepted: 01/27/2006] [Indexed: 10/25/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine with important roles in inflammation, wound repair, and cancer. Cells secrete TGF-beta as a latent protein complex, consisting of disulfide-bonded homodimers of growth factor and latency-associated propeptide. Latency regulates extracellular TGF-beta action by controlling the levels of active growth factor available. We report here that active and latent TGF-beta were inactivated in vitro by reduction of the growth factor dimer under physiological conditions. We also demonstrate that the latency-associated propeptide has chaperone-like activity and partially protects TGF-beta from inactivation. TGF-beta inactivation occurred upon incubation with the physiological redox agents, cysteine, homocysteine, and reduced glutathione. Inactivation was temperature- and dose-dependent. While inactivation by physiological concentrations of redox agents was partial at 37 degrees C, active and latent TGF-beta were completely inactivated by raising the temperature in the presence of the redox agents. The mechanism of TGF-beta inactivation involved the generation of biologically inactive growth factor monomer and required the presence of free thiol groups, since thiol blockers protected TGF-beta from reduction. We conclude that non-enzymatic redox reactions may be involved in the regulation of extracellular TGF-beta activity. This might be of particular relevance in wound repair (e.g. in burns), as a mechanism protecting from excess TGF-beta activity, as well as in conditions involving redox dysregulation, such as reperfusion injury of the heart, Alzheimer's disease, and cancer.
Collapse
Affiliation(s)
- Robert Blakytny
- Department of Cancer Research, Fachklinik Hornheide, D-48157 Münster, Germany.
| | | | | |
Collapse
|
22
|
Ramanathan B, Jan KY, Chen CH, Hour TC, Yu HJ, Pu YS. Resistance to paclitaxel is proportional to cellular total antioxidant capacity. Cancer Res 2005; 65:8455-60. [PMID: 16166325 DOI: 10.1158/0008-5472.can-05-1162] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Paclitaxel, one of the most commonly prescribed chemotherapeutic agents, is active against a wide spectrum of human cancer. The mechanism of its cytotoxicity, however, remains controversial. Our results indicate that paclitaxel treatment increases levels of superoxide, hydrogen peroxide, nitric oxide (NO), oxidative DNA adducts, G2-M arrest, and cells with fragmented nuclei. Antioxidants pyruvate and selenium, the NO synthase inhibitor N(omega)-nitro-L-arginine methyl ester, and the NO scavenger manganese (III) 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide all decreased paclitaxel-mediated DNA damage and sub-G1 cells. In contrast, the glutamylcysteine synthase inhibitor buthionine sulfoximine (BSO) and the superoxide dismutase (SOD) inhibitor 2-methoxyestradiol (2-ME) increased the sub-G1 fraction in paclitaxel-treated cells. These results suggest that reactive oxygen and nitrogen species are involved in paclitaxel cytotoxicity. This notion is further supported with the observation that concentrations of paclitaxel required to inhibit cell growth by 50% correlate with total antioxidant capacity. Moreover, agents such as arsenic trioxide (As2O3), BSO, 2-ME, PD98059, U0126 [mitogen-activated protein/extracellular signal-regulated kinase inhibitors], and LY294002 (phosphatidylinositol 3-kinase/Akt inhibitor), all of which decrease clonogenic survival, also decrease the total antioxidant capacity of paclitaxel-treated cells, regardless whether they are paclitaxel sensitive or paclitaxel resistant. These results suggest that paclitaxel chemosensitivity may be predicted by taking total antioxidant capacity measurements from clinical tumor samples. This, in turn, may then improve treatment outcomes by selecting out potentially responsive patients.
Collapse
|
23
|
Kim J, Bae SM, Lim DS, Kwak SY, Lee CK, Lee YS, Bae IJ, Yoo JY, Lee YJ, Kim CK, Ahn WS. Tetraarsenic oxide-mediated apoptosis in a cervical cancer cell line, SiHa. Cancer Res Treat 2005; 37:307-12. [PMID: 19956532 DOI: 10.4143/crt.2005.37.5.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Accepted: 09/21/2005] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Diarsenic oxide, As(2)O(3), has been reported to be effective in treating acute leukemia, and induce apoptosis in many tumor cells. In this study, the ability of a novel arsenical compound, As(4)O(6) (tetraarsenic oxide), along with As(2)O(3), for its ability to induce cell growth inhibition, as well as apoptosis, in human cervical cancer cells, SiHa cells, were evaluated in vitro. MATERIALS AND METHODS To examine the levels of apoptosis, SiHa cells were given two sensitive doses, 0.5 and 1 microM, of arsenical compounds, and a DNA fragmentation assay and FACS analysis were then conducted. In addition, a Western blotting assay was performed to identify target molecules for apoptosis. RESULTS Both As(2)O(3) and As(4)O(6) induced dosedependent inhibition of SiHa cell proliferation. In particular, As(4)O(6) was more effective at suppressing SiHa cell growth than As(2)O(3). In parallel with the inhibition of cell proliferation, As(4)O(6) caused a significantly greater increase in the sub-G1 cell population than As(2)O(3), as determined by propidium iodide DNA staining. This was confirmed by a DNA fragmentation assay and annexin V staining. The Western blotting analysis also showed that the expression of proliferating cell nuclear antigen (PCNA) was suppressed to a significantly greater extent by As(4)O(6) than As(2)O(3) at a dose of 0.5 microM. However, the apoptosis-related protein, Bax, was expressed to a significantly greater extent due to As(4)O(6) than As(2)O(3). CONCLUSION Taken together, these findings suggest that a novel arsenic compound, As(4)O(6), possesses more potent anti-proliferative effects on human cervical cancer cells, with the induction of apoptosis also, at least via the activation of Bax protein in vitro.
Collapse
Affiliation(s)
- Jeong Kim
- Departments of Obstetrics and Gynecology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The proven efficacy of ATO in the treatment of APL and the emerging importance of ATO in other diseases prompted extensive studies of the mechanisms of action of ATO in APL and in other types of cancers. In this review we will focus on downstream events in ATO-induced intrinsic and extrinsic apoptotic pathways with an emphasis on the role of pro-apoptotic and anti-apoptotic proteins and the role of p53 in ATO-induced apoptosis including its effect on cell cycle, its anti-mitotic effect and the role of apoptosis inducing factors (AIF) in ATO-induced apoptosis, chromatin condensation and nuclear fragmentation in myeloma cells as a model.
Collapse
Affiliation(s)
- Yair Gazitt
- Department of Medicine/Hematology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78284, USA.
| | | |
Collapse
|
25
|
Chou WC, Dang CV. Acute promyelocytic leukemia: recent advances in therapy and molecular basis of response to arsenic therapies. Curr Opin Hematol 2005; 12:1-6. [PMID: 15604884 DOI: 10.1097/01.moh.0000148552.93303.45] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW While arsenic has long been known as a poison and environmental carcinogen, its dramatic effect in the treatment of acute promyelocytic leukemia (APL) has made its mechanism of action a topic of intense interest. This paper reviews recent findings that reveal why a traditional poison has become a magical potion for a major type of APL, which is characterized by a balanced chromosomal translocation t(15;17). RECENT FINDINGS Daily IV infusion of arsenic trioxide (As2O3; ATO) for 30 to 40 days can lead to complete remission in about 85% of patients with newly diagnosed or relapsed APL. Oral preparations of ATO and tetra-arsenic tetra-sulfide (As4S4) seem to be as effective as parenteral ATO, with similar toxicity profiles. The combination of all-trans retinoic acid and ATO in patients with newly diagnosed APL has yielded more durable remission than monotherapy. The mechanism of arsenic cytotoxicity is thought to involve posttranslational modification followed by degradation of the PML-retinoic acid receptor-alpha (PML-RARalpha) fusion protein; targeting of PML to nuclear bodies with restoration of its physiologic functions; and production of reactive oxygen species (ROS) by NADPH oxidase in leukemic cells or collapse of the mitochondrial transmembrane potential. The understanding of arsenic cytotoxicity has stimulated modifications that promise to improve efficacy, such as interfering with ROS scavenging or boosting of ROS production to enhance the cytotoxicity, and adding cAMP or interferons to ATO regimens. SUMMARY Recent advances in the clinical use of arsenic, the mechanism of arsenic-mediated cytotoxicity, and modulations of ATO to increase its efficacy and expand its clinical spectrum are reviewed.
Collapse
Affiliation(s)
- Wen-Chien Chou
- Department of Laboratory Medicine and Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | |
Collapse
|
26
|
Rey NA, Howarth OW, Pereira-Maia EC. Equilibrium characterization of the As(III)-cysteine and the As(III)-glutathione systems in aqueous solution. J Inorg Biochem 2005; 98:1151-9. [PMID: 15149827 DOI: 10.1016/j.jinorgbio.2004.03.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2003] [Revised: 03/19/2004] [Accepted: 03/24/2004] [Indexed: 11/25/2022]
Abstract
Some arsenic compounds were the first antimicrobial agents specifically synthesized for the treatment of infectious diseases such as syphilis and trypanosomiasis. More recently, arsenic trioxide has been shown to be efficient in the treatment of acute promyelocytic leukemia. The exact mechanism of action has not been elucidated yet, but it seems to be related to arsenic binding to vicinal thiol groups of regulatory proteins. Glutathione is the major intracellular thiol and plays important roles in the cellular defense and metabolism. This paper reports on a study of the interactions between arsenic(III) and either cysteine or glutathione in aqueous solution. The behavior observed for the As(III)-glutathione system is very similar to that of As(III)-cysteine. In both cases, the formation of two complexes in aqueous solution was evidenced by NMR and electronic spectroscopies and by potentiometry. The formation constants of the cysteine complexes [As(H(-1)Cys)(3)], log K = 29.84(6), and [As(H(-2)Cys)(OH)(2)](-), log K = 12.01(9), and of the glutathione complexes [As(H(-2)GS)(3)](3-), log K = 32.0(6), and [As(H(-3)GS)(OH)(2)](2-), log K = 10(3) were calculated from potentiometric and spectroscopic data. In both cases, the [As(HL)(3)] species, in which the amine groups are protonated, predominate from acidic to neutral media, and the [As(L)(OH)(2)] species appear in basic medium (the charges were omitted for the sake of simplicity). Spectroscopic data clearly show that the arsenite-binding site in both complexes is the sulfur atom of cysteine. In the [As(L)(OH)(2)] species, the coordination sphere is completed by two hydroxyl groups. In both cases, arsenic probably adopts a trigonal pyramidal geometry. Above pH 10, the formation of [As(OH)(2)O](-) excludes the thiolates from arsenic coordination sites. At physiological pH, almost 80% of the ligand is present as [As(HL)(3)].
Collapse
Affiliation(s)
- Nicolás A Rey
- Departamento de Química - ICEx, Universidade Federal de Minas Gerais 31.270-901 Belo Horizonte, Minas Gerais, Brazil
| | | | | |
Collapse
|
27
|
Lau A, He QY, Chiu JF. A proteome analysis of the arsenite response in cultured lung cells: evidence for in vitro oxidative stress-induced apoptosis. Biochem J 2004; 382:641-50. [PMID: 15175009 PMCID: PMC1133821 DOI: 10.1042/bj20040224] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Revised: 05/10/2004] [Accepted: 06/03/2004] [Indexed: 01/16/2023]
Abstract
Arsenite is well documented as a chemotherapeutic agent capable of inducing cell death. However, the cellular response at the molecular level has not been studied extensively. In the present study, we provide for the first time a proteomic analysis of rat LECs (lung epithelial cells) treated with arsenite, with the aim of identifying defence proteins, probably expressed to protect the cells during the course of arsenic-induced apoptosis. Comparative proteome analysis was conducted on LECs and LECs treated with 40 microM arsenite to identify global changes in their protein expression profiles. Over 1000 protein spots were separated by two-dimensional electrophoresis and visualized by silver staining. Seven proteins changed expression levels significantly and were identified by matrix-assisted laser-desorption ionization-time-of-flight mass spectrometry and database searching. The proteins up-regulated were mostly HSPs (heat-shock proteins) and antioxidative stress proteins, including HSP70, aldose reductase, haem oxygenase-1, HSP27, ferritin light chain and alphaB-crystallin. The glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase was down-regulated. Pretreatment with the thiol antioxidants glutathione or N-acetylcysteine before arsenite insult effectively abrogated the induction of these defence proteins and sustained cell viability, whereas antioxidants were protective only at earlier time points if they were added to cells after arsenite. Taken together, our results demonstrate that high levels of arsenite cause oxidative stress-induced apoptosis.
Collapse
Key Words
- apoptosis
- arsenite
- heat shock protein (hsp)
- matrixassisted laser-desorption ionization–time-of-flight mass spectrometry (maldi–tof-ms)
- oxidative stress
- reactive oxygen species (ros)
- αb-c, αb-crystallin
- ar, aldose reductase
- dapi, 4,6-diamidino-2-phenylindole
- 2-de, two-dimensional electrophoresis
- flc, ferritin light chain
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- ho-1, haem oxygenase-1
- hsp, heat-shock protein
- shsp, small heat shock or stress protein
- ief, isoelectric focusing
- jnk, c-jun n-terminal kinase
- lec, lung epithelial cell
- maldi–tof-ms, matrix-assisted laser-desorption ionization–time-of-flight mass spectrometry
- nac, n-acetylcysteine
- ros, reactive oxygen species
Collapse
Affiliation(s)
- Andy T. Y. Lau
- *Institute of Molecular Biology, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
- †Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
| | - Qing-Yu He
- †Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
- ‡Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
| | - Jen-Fu Chiu
- *Institute of Molecular Biology, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
- †Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People's Republic of China
| |
Collapse
|
28
|
Kim YW, Bae SM, Lee KH, Lee JM, Namkoong SE, Lee IP, Kim CK, Seo JS, Sin JI, Kim YW, Ahn WS. Comparison of As(2)O(3) and As(4)O(6) in the detection of SiHa cervical cancer cell growth inhibition pathway. Cancer Res Treat 2004; 36:255-62. [PMID: 20368843 DOI: 10.4143/crt.2004.36.4.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2004] [Accepted: 08/13/2004] [Indexed: 11/21/2022] Open
Abstract
PURPOSE An arsenical compound, As(2)O(3), has been reported to be effective for treating acute leukemia and inducing apoptosis in many different tumor cells. In this study, the ability of As(4)O(6) to suppress cell growth and induce gene expression patterns was tested using a cDNA microarray in HPV16 immortalized cervical carcinoma cells, SiHa cells, along with As(2)O(3). MATERIALS AND METHODS A novel arsenical compound, As(4)O(6), was designed and its ability to induce cell growth inhibition as well as gene expression profiles along with As(2)O(3) in HPV16 infected SiHa cervical cancer cells was compared. Both As(2)O(3) and As(4)O(6) induced apoptosis in SiHa cells, as determined by DNA ladder formation. To further compare the gene expression profiles between these two drugs, a 384 cDNA microarray system was employed. Also, the gene expression profiles were classified into the Gene Ontology (GO) to investigate apoptosis-related cellular processes. RESULTS As(4)O(6) was more effective i suppressing the growth of SiHa cells in vitro compared to As(2)O(3). In the case of treatment with As(2)O(3), 41 genes were up- or down-regulated at least 2 fold compared to non-treatment. However, 65 genes were up- or down-regulated by As(4)O(6) treatment. In particular, 27 genes were commonly regulated by both arsenic compounds. Also, the GO analysis indicated that down-regulation of cell-regulatory functions, such as cell cycle, protein kinase activity and DNA repair, induced anti-tumor effect. CONCLUSION These data support that As(4)O(6) could be more effective than As(2)O(3) in inhibiting the growth of HPV16 infected cervical cancer cells. This appears to be mediated through a unique, but overlapping regulatory mechanism(s), suggesting that the regulated genes and cellular processes could be further used as a new potential drug approach for treating cervical cancer in clinical settings.
Collapse
Affiliation(s)
- Yong Wook Kim
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lu M, Xia L, Luo D, Waxman S, Jing Y. Dual effects of glutathione-S-transferase π on As2O3 action in prostate cancer cells: enhancement of growth inhibition and inhibition of apoptosis. Oncogene 2004; 23:3945-52. [PMID: 15007384 DOI: 10.1038/sj.onc.1207500] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To determine the effects of glutathione-S-transferase pi (GSTpi) on the actions of As2O3, As2O3-induced growth inhibition and apoptosis was studied in three prostate cancer cell lines: DU-145, PC-3 and LNCaP cells. As2O3 inhibited cell proliferation of DU-145 and PC-3 cells (both cells express GSTpi), but not of LNCaP cells (which lack GSTpi expression) at concentrations below 1 microM. LNCaP cells stably transfected and expressed GSTpi (LNCaP/GSTpi) became sensitive to As2O3 growth inhibition. As2O3 arrested cell growth of DU-145, PC-3 and LNCaP/GSTpi cells in the G2/M phase of the cell cycle at low concentrations (<2 microM), but did not induce apoptosis. At higher concentrations (10-20 microM), As2O3 induced apoptosis in LNCaP cells, but not in DU-145 or PC-3 cells. The apoptosis induction due to As2O3 treatment of LNCaP cell correlated with the activation of JNK and p38 and induction of p53 protein. LNCaP/GSTpi cells became insensitive to As2O3-induced apoptosis with reduced JNK activition. These data indicate that GSTpi increases growth inhibition due to As2O3 treatment and prevents As2O3-induced apoptosis in prostate cancer cells. Therefore, it appears that As2O3 inhibits cell growth and induces apoptosis through different mechanisms.
Collapse
Affiliation(s)
- Min Lu
- Division of Hematology/Oncology, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
30
|
Hirano S, Cui X, Li S, Kanno S, Kobayashi Y, Hayakawa T, Shraim A. Difference in uptake and toxicity of trivalent and pentavalent inorganic arsenic in rat heart microvessel endothelial cells. Arch Toxicol 2003; 77:305-12. [PMID: 12799770 DOI: 10.1007/s00204-003-0447-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2002] [Accepted: 12/18/2002] [Indexed: 11/30/2022]
Abstract
Intake of inorganic arsenic is known to cause vascular diseases as well as skin lesions and cancer in humans. We investigated the differences in cytotoxicity, uptake rate of arsenic, and gene expression of antioxidative enzymes between arsenite (As(3+))- and arsenate (As(5+))-exposed rat heart microvessel endothelial cells. As(3+) was more cytotoxic than As(5+), and LC(50) values were calculated to be 36 and 220 micro M, respectively. As(3+) (1-25 micro M) increased mRNA levels of antioxidant enzymes such as heme oxygenase-1 (HO-1), thioredoxin peroxidase 2, NADPH dehydrogenase, and glutathione S-transferase P subunit. HO-1 mRNA levels showed the most remarkable increase in response to As(3+). cDNA microarray analysis indicated that there was no prominent difference in arsenic-induced transcriptional changes between As(3+)- and As(5+)-exposed cells, when the cells were exposed to one-fourth the LC(50) concentration of arsenic (9 and 55 micro M for As(3+) and As(5+), respectively). N-acetyl- l-cysteine (NAC) reduced both the cytotoxicity of inorganic arsenic and the HO-1 mRNA level, and buthionine sulfoximine enhanced cytotoxicity of inorganic arsenic. As(3+) was taken up by the endothelial cells 6-7 times faster than As(5+), and the presence of NAC in the culture medium did not change the uptake rate of As(3+). These results suggest that the effects of NAC on arsenic-induced cytotoxicity and oxidative stress were due to the antioxidative role of non-protein thiols and not to chelation of arsenic in the culture medium. The difference in cellular uptake of arsenic between As(3+) and As(5+) appeared not to be due to the ionic charge on arsenic (at physiological pH, trivalent arsenic is neutral whereas pentavalent arsenic is negatively charged). These results suggest that the higher toxicity of As(3+) compared with that of As(5+) is probably due to the faster uptake of As(3+) by endothelial cells, and inorganic arsenic exerts its toxicity at least in part via intracellular oxidative stress.
Collapse
Affiliation(s)
- Seishiro Hirano
- Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, 305-8506, Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | | | |
Collapse
|
31
|
Ling YH, Jiang JD, Holland JF, Perez-Soler R. Arsenic trioxide produces polymerization of microtubules and mitotic arrest before apoptosis in human tumor cell lines. Mol Pharmacol 2002; 62:529-38. [PMID: 12181429 DOI: 10.1124/mol.62.3.529] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Arsenic trioxide (As(2)O(3)) has been found to induce apoptosis in leukemia cell lines and clinical remissions in patients with acute promyelocytic leukemia. In this study, we investigated the cytotoxic effect and mechanisms of action of As(2)O(3) in human tumor cell lines. As(2)O(3) caused inhibition of cell growth (IC(50) range, 3-14 microM) in a variety of human solid tumor cell lines, including four human non-small-cell lung cancer cell lines (H460, H322, H520, H661), two ovarian cancer cell lines (SK-OV-03, A2780), cervical cancer HeLa, and breast carcinoma MCF-7, as assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Flow cytometry analysis showed that As(2)O(3) treatment resulted in a time-dependent accumulation of cells in the G(2)/M phase. We observed, using Wright-Giemsa and 4',6-diamidine-2-phenylindole-dihydrochloride staining, that As(2)O(3) blocked the cell cycle in mitosis. In vitro examination revealed that As(2)O(3) markedly promoted tubulin polymerization without affecting GTP binding to beta-tubulin. Immunocytochemical and EM studies of treated MCF-7 cells showed that As(2)O(3) treatment caused changes in the cellular microtubule network and formation of polymerized microtubules. Similar to most anti-tubulin agents, As(2)O(3) treatment induced up-regulation of the cyclin B1 levels and activation of p34(cdc2)/cyclinB1 kinase, as well as Bcl-2 phosphorylation. Furthermore, activation of caspase-3 and -7 and cleavage of poly(ADP-ribose) polymerase and beta-catenin occurred only in As(2)O(3)-induced mitotic cells, not in interphase cells, suggesting that As(2)O(3)-induced mitotic arrest may be a requirement for the activation of apoptotic pathways. In addition, As(2)O(3) exhibited similar inhibitory effects against parental MCF-7, P-glycoprotein-overexpressing MCF-7/doxorubicin cells, and multidrug resistance protein (MRP)-expressing MCF-7/etoposide cells (resistance indices, 2.3 and 1.9, respectively). Similarly, As(2)O(3) had similar inhibitory effect against parental ovarian carcinoma A2780 cells and tubulin mutation paclitaxel-resistant cell lines PTx10 and PTx22 (resistance indices, 0.86 and 0.93, respectively), suggesting that its effect on tubulin polymerization and G(2)/M phase arrest is distinct from that of paclitaxel. Taken together, our data demonstrate that As(2)O(3) has a paclitaxel-like effect, markedly promotes tubulin polymerization, arrests cell cycle at mitosis, and induces apoptosis. In addition, As(2)O(3) is a poor substrate for transport by P-glycoprotein and MRP, and non-cross-resistant with paclitaxel resistant cell lines due to tubulin mutation, suggesting that As(2)O(3) may be useful for treatment of human solid tumors, particularly in patients with paclitaxel resistance.
Collapse
Affiliation(s)
- Yi-He Ling
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | | | |
Collapse
|
32
|
Chen A, Cao EH, Zhang TC, Qin JF. Arsenite-induced reactive oxygen species and the repression of alpha-tocopherol in the MGC-803 cells. Eur J Pharmacol 2002; 448:11-8. [PMID: 12126965 DOI: 10.1016/s0014-2999(02)01901-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We have investigated the action of oxidative stress in arsenite-induced apoptosis of human gastric cancer MGC-803 cells. Cells exhibited obvious characteristic of apoptosis following the treatment with 1.0 microM arsenite for 24 h. During the process, low concentration of arsenite significantly increased superoxide formation and lipid peroxidation, which was dose-dependent and was related to cell apoptosis induced by arsenite. The oxidant-dependent increase in intracellular [Ca(2+)] level and p53 gene expression were also observed at the same time. A phospholipase C inhibitor, 1-[6-([(17 beta)-3-methoxyestra-1,3,5,(10)-trien-17-yl]-amino)hexyl]-2,5-dione (U73122), could block the rapid transient increase in intracellular Ca(2+) levels, as well as the subsequent fragmentation of nuclear DNA. Addition of alpha-tocopherol before arsenite treatment abolished the transient increase in superoxide formation, lipid peroxidation, intracellular [Ca(2+)] levels and p53 gene expression, and furthermore could significantly inhibited the arsenite-induced apoptosis of MGC-803 cells. These results indicate that arsenite-induced oxidative stress, which stimulate cellular signaling systems, are involved in apoptosis of MGC-803 cells.
Collapse
Affiliation(s)
- Ai Chen
- Institute of Biophysics, Acadenia Sinica, 15 Datun Road, Chaoyang District, Beijing 100101, PR China
| | | | | | | |
Collapse
|
33
|
Abstract
Two clonal nerve-like cell lines derived from HT22 and PC12 have been selected for resistance to glutamate toxicity and amyloid toxicity, respectively. In the following experiments it was asked if these cell lines show cross-resistance toward amyloid beta peptide (Abeta) and glutamate as well as toward a variety of additional neurotoxins. Conversely, it was determined if inhibitors of oxytosis, a well-defined oxidative stress pathway, also protect cells from the neurotoxins. It is shown that both glutamate and amyloid resistant cells are cross resistant to most of the other toxins or toxic conditions, while inhibitors of oxytosis protect from glutathione and cystine depletion and H2O2 toxicity, but not from the toxic effects of nitric oxide, rotenone, arsenite or cisplatin. It is concluded that while there is a great deal of cross-resistance to neurotoxins, the components of the cell death pathway which has been defined for oxytosis are not used by many of the neurotoxins.
Collapse
Affiliation(s)
- Richard Dargusch
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | |
Collapse
|
34
|
Bode AM, Dong Z. The paradox of arsenic: molecular mechanisms of cell transformation and chemotherapeutic effects. Crit Rev Oncol Hematol 2002; 42:5-24. [PMID: 11923065 DOI: 10.1016/s1040-8428(01)00215-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Arsenic is a well-documented carcinogen that also appears to be a valuable therapeutic tool in cancer treatment. This creates a paradox for which no unified hypothesis has been reached regarding the molecular mechanisms that determine whether arsenic will act as a carcinogen or as an effectual chemotherapeutic agent. Much of our knowledge with respect to the actions of arsenic has been drawn from epidemiological or clinical studies. The actions of arsenic are likely to be related to cell type, arsenic species, and length and dose of exposure. Arsenic unquestionably induces apoptosis and may specifically target certain tumor cells. Research data strongly suggest that arsenic influences distinct signaling pathways involved in mediating proliferation or apoptosis, including mitogen-activated protein kinases, p53, activator protein-1 or nuclear factor kappa B. The primary purpose of this review is to examine recent findings, from this laboratory and others, that focus on the molecular mechanisms of arsenic's actions in cell transformation and as a therapeutic agent.
Collapse
Affiliation(s)
- Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | | |
Collapse
|
35
|
Abstract
Caspases are critical mediators of apoptotic cell death. All members of the caspase family contain the sequence QACXG which contains the active site cysteine. The putative active site of caspase 3 contains a cysteine residue that is subject to redox control. Both thioredoxin and glutathione have been shown to be required for caspase-3 activity to induce apoptosis. The regulation of inducible caspase 3 activity by oxidation-reduction (redox) dependent mechanisms is reviewed. Up until a few years ago, reactive oxygen species (ROS) research mostly focussed on oxidative damage and ROS were thought to be a key trigger for cell death. This view has been refined, leading to the understanding that the biological function of ROS is determined by numerous variables such as concentration, chemical type and cellular localization. For example, ROS and reactive nitrogen species may intercept inducible cell death under certain circumstances via the redox regulation of inducible caspase activity and/or by depleting cellular energy stores. Likewise, death of unwanted diseased or degenerative cells may be facilitated by pharmacologically enhancing the thiol status of such cells using redox-active α-lipoic acid.
Collapse
Affiliation(s)
- C K Sen
- Laboratory of Molecular Medicine, Department of Surgery, 512 Heart and Lung Research Institute, The Ohio State University Medical Center, 473 W. 12th Avenue, Columbus, OH 43210, USA
| | | |
Collapse
|
36
|
Wang TS, Hsu TY, Chung CH, Wang AS, Bau DT, Jan KY. Arsenite induces oxidative DNA adducts and DNA-protein cross-links in mammalian cells. Free Radic Biol Med 2001; 31:321-30. [PMID: 11461769 DOI: 10.1016/s0891-5849(01)00581-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Arsenic is generally recognized as a nonmutagenic carcinogen because sodium arsenite induces DNA damage only at very high concentrations. In this study we demonstrate that arsenite concentrations above 0.25 microM induce DNA strand breaks in both human leukemia cells and Chinese hamster ovary cells. Therefore, DNA damage may be involved in arsenic-induced carcinogenesis. Formamidopyrimidine-DNA glycosylase and proteinase K greatly increased DNA strand breaks in arsenite-treated cells, providing evidence that a large portion of arsenite-induced DNA strand breaks come from excision of oxidative DNA adducts and DNA-protein cross-links. Because DNA strand breaks appear only temporarily during excision repair, the level of detectable DNA strand breaks will be low at any given time point. For this reason many previous studies have only detected low levels of DNA strand breaks. We also show that catalase, and inhibitors of calcium, nitric oxide synthase, superoxide dismutase, and myeloperoxidase, could modulate arsenite-induced DNA damage. We conclude that arsenite induces DNA adducts through calcium-mediated production of peroxynitrite, hypochlorous acid, and hydroxyl radicals.
Collapse
Affiliation(s)
- T S Wang
- Department of Life Science, Chung Shan Medical and Dental College, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Treatment of acute promyelocytic leukemia with arsenic compounds: In vitro and in vivo studies. Semin Hematol 2001. [DOI: 10.1016/s0037-1963(01)90003-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Tartier L, McCarey YL, Biaglow JE, Kochevar IE, Held KD. Apoptosis induced by dithiothreitol in HL-60 cells shows early activation of caspase 3 and is independent of mitochondria. Cell Death Differ 2000; 7:1002-10. [PMID: 11279547 DOI: 10.1038/sj.cdd.4400726] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Previous studies have shown that under certain conditions some thiol-containing compounds can cause apoptosis in a number of different cell lines. Herein, we investigated the apoptotic pathways in HL-60 cells triggered by dithiothreitol (DTT), used as a model thiol compound, and tested the hypothesis that thiols cause apoptosis via production of hydrogen peroxide (H2O2) during thiol oxidation. The results show that, unlike H2O2, DTT does not induce apoptosis via a mitochondrial pathway. This is demonstrated by the absence of early cytochrome c release from mitochondria into the cytosol, the lack of mitochondrial membrane depolarization at early times, and the minor role of caspase 9 in DTT-induced apoptosis. The first caspase activity detectable in DTT-treated cells is caspase 3, which is increased significantly 1 - 2 h after the start of DTT treatment. This was shown by following the cleavage of both a natural substrate, DFF-45/ICAD, and a synthetic fluorescent substrate, z-DEVD-AFC. Cleavage of substrates of caspases 2 and 8, known as initiator caspases, does not start until 3 - 4 h after DTT exposure, well after caspase 3 has become active and at a time when apoptosis is in late stages, as shown by the occurrence of DNA fragmentation to oligonucleosomal-sized pieces. Although oxidizing DTT can produce H2O2, data presented here indicate that DTT-induced apoptosis is not mediated by production of H2O2 and occurs via a novel pathway that involves activation of caspase 3 at early stages, prior to activation of the common 'initiator' caspases 2, 8 and 9.
Collapse
Affiliation(s)
- L Tartier
- Laboratory of Molecular and Cellular Radiation Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
39
|
Cai X, Shen YL, Zhu Q, Jia PM, Yu Y, Zhou L, Huang Y, Zhang JW, Xiong SM, Chen SJ, Wang ZY, Chen Z, Chen GQ. Arsenic trioxide-induced apoptosis and differentiation are associated respectively with mitochondrial transmembrane potential collapse and retinoic acid signaling pathways in acute promyelocytic leukemia. Leukemia 2000; 14:262-70. [PMID: 10673743 DOI: 10.1038/sj.leu.2401650] [Citation(s) in RCA: 197] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent studies showed that arsenic trioxide (As2O3) could induce apoptosis and partial differentiation of leukemic promyelocytes. Here, we addressed the possible mechanisms underlying these two different effects. 1.0 microM As2O3-induced apoptosis was associated with condensation of the mitochondrial matrix, disruption of mitochondrial transmembrane potentials (DeltaPsim) and activation of caspase-3 in acute promyelocytic leukemia (APL) cells regardless of their sensitivity to all-trans retinoic acid (ATRA). All these effects were inhibited by dithiothreitol (DTT) and enhanced by buthionine sulfoximine (BSO). Furthermore, BSO could also render HL60 and U937 cells, which had the higher cellular catalase activity, sensitive to As2O3-induced apoptosis. Surprisingly, 1.0 microM As2O3 did not induce the DeltaPsim collapse and apoptosis, while 0.1 microM As2O3 induced partial differentiation of fresh BM cells from a de novo APL patient. In this study, we also showed that 0.2 mM DTT did not block low-dose As2O3-induced NB4 cell differentiation, and 0. 10.5 microM As2O3 did not induce differentiation of ATRA-resistant NB4-derived sublines, which were confirmed by cytomorphology, expression of CD11b, CD33 and CD14 as well as NBT reduction. Another interesting finding was that 0.10.5 microM As2O3 could also induce differentiation-related changes in ATRA-sensitive HL60 cells. However, the differentiation-inducing effect could not be seen in ATRA-resistant HL60 sublines with RARalpha mutation. Moreover, low-dose As2O3 and ATRA yielded similar gene expression profiles in APL cells. These results encouraged us to hypothesize that As2O3 induces APL cell differentiation through direct or indirect activation of retinoic acid receptor-related signaling pathway(s), while DeltaPsim collapse is the common mechanism of As2O3-induced apoptosis.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/pharmacology
- Caspase 3
- Caspases/metabolism
- Cell Differentiation/drug effects
- DNA, Neoplasm/analysis
- Electrophoresis, Agar Gel
- Enzyme Precursors/metabolism
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- HL-60 Cells
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/enzymology
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Membrane Potentials/drug effects
- Microscopy, Electron
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mutation
- Oxides/pharmacology
- Receptors, Retinoic Acid/genetics
- Retinoic Acid Receptor alpha
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/drug effects
- Tretinoin/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- X Cai
- Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Second Medical University, 197 Rui-Jin Road II, Shanghai, 200025, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bode A, Dong Z. Apoptosis induction by arsenic: mechanisms of action and possible clinical applications for treating therapy-resistant cancers. Drug Resist Updat 2000; 3:21-29. [PMID: 11498362 DOI: 10.1054/drup.2000.0114] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Arsenic, a known carcinogen, may be useful in cancer treatment. Arsenic may be effective in counteracting drug resistance because it appears to induce apoptosis in tumor cells independently of p53 activation, thereby allowing it to be directed against p53-defective cancers. The role of MAP kinases in arsenic-induced apoptosis in tumor cells is important and may be influenced by reactive oxygen species or glutathione. This review focuses on recent findings from this and other laboratories regarding the mechanism(s) of arsenic-induced apoptosis in tumor cells and considers their relevance in the clinical treatment of therapy-resistant cancers. Copyright 2000 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Ann Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | | |
Collapse
|
41
|
Abstract
Reactive oxygen species have been shown to be involved in the mutagenicity, clastogenicity, and apoptosis of mammalian cells treated with arsenic or cadmium. As these endpoints require several hours of cellular processing, it is not clear that reactive oxygen species damage DNA directly or interfere with DNA replication and repair. Using single-cell alkaline electrophoresis, we have detected DNA strand breaks (DSBs) in bovine aortic endothelial cells by a 4-h treatment with sodium arsenite (As) and cadmium chloride (Cd) in sublethal concentrations. As-induced DSBs could be decreased by nitric oxide (NO) synthase inhibitors, superoxide scavengers, and peroxynitrite scavengers and could be increased by superoxide generators and NO generators. Treatment with As also increased nitrite production. These results suggest that As-increased NO may react with O2*- to produce peroxynitrite and cause DNA damage. The results showing that Cd increased cellular H2O2 levels and that Cd-induced DSBs could be modulated by various oxidant modulators suggest that Cd may induce DSBs via O2*-, H2O2, and *OH. Nevertheless, the DSBs in both As- and Cd-treated cells seem to come from the excision of oxidized bases such as formamidopyrimidine and 8-oxoguanine, as the Escherichia coli enzyme formamidopyrimidine-DNA glycosylase (Fpg) increased DSBs in cells treated with As, 3-morpholinosydnonimine (a peroxynitrite-generating agent), Cd, or H2O2.
Collapse
Affiliation(s)
- F Liu
- Institute of Zoology, Academia Sinica, Taipei, Taiwan, Republic of China
| | | |
Collapse
|