1
|
El-Mahrouk SR, El-Ghiaty MA, Alqahtani MA, El-Kadi AOS. Arsenic Trioxide (ATO III) Induces NAD(P)H Quinone Oxidoreductase 1 (NQO1) Expression in Hepatic and Extrahepatic Tissues of C57BL/6 Mice. Chem Res Toxicol 2024; 37:2040-2051. [PMID: 39630573 DOI: 10.1021/acs.chemrestox.4c00422] [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: 12/07/2024]
Abstract
Arsenic trioxide (ATOIII) has emerged as a potent therapeutic agent for acute promyelocytic leukemia (APL), yet its clinical application is often limited by significant adverse effects. This study investigates the molecular mechanisms underlying ATOIII's impact on cellular detoxification pathways, focusing on the regulation of NAD(P)H/quinone oxidoreductase (NQO1), a crucial enzyme in maintaining cellular homeostasis and cancer prevention. We explored ATOIII's effects on NQO1 expression in C57BL/6 mice and Hepa-1c1c7 cells, both independently and in combination with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a known NQO1 inducer. Our findings revealed that ATOIII significantly increased NQO1 expression in hepatic and extrahepatic tissues, as well as in Hepa-1c1c7 cells, at mRNA, protein, and activity levels. This upregulation occurred both in the presence and absence of TCDD. Mechanistically, we demonstrated that ATOIII promotes the nuclear translocation of both nuclear factor erythroid 2-related factor-2 (NRF2) and aryl hydrocarbon receptor (AHR) transcription factors. Furthermore, ATOIII exposure increased antioxidant response element (ARE)-driven reporter gene activity, indicating a transcriptional mechanism of NQO1 induction. Notably, gene silencing experiments confirmed the critical roles of both NRF2 and AHR in mediating ATOIII-induced NQO1 expression. In conclusion, ATOIII exposure is found to upregulate the NQO1 enzyme through a transcriptional mechanism via AHR- and NRF2- dependent mechanisms, offering valuable insights into its therapeutic mechanisms.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Faculty of Pharmacy, Tanta University, Gharbia, Tanta 31111, Egypt
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| |
Collapse
|
2
|
Lu J, Chen L, Fatima Z, Huang J, Chen J. Synergistic rescue of temperature-sensitive p53 mutants by hypothermia and arsenic trioxide. Mol Carcinog 2024; 63:2205-2217. [PMID: 39115446 PMCID: PMC11466696 DOI: 10.1002/mc.23804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/29/2024] [Indexed: 10/11/2024]
Abstract
The p53 tumor suppressor is inactivated by mutations in about 50% of tumors. Rescuing the transcriptional function of mutant p53 has potential therapeutic benefits. Approximately 15% of p53 mutants are temperature sensitive (TS) and regain maximal activity at 32°C. Proof of concept study showed that induction of 32°C hypothermia in mice restored TS mutant p53 activity and inhibited tumor growth. However, 32°C is the lower limit of therapeutic hypothermia procedures for humans. Higher temperatures are preferable but result in suboptimal TS p53 activation. Recently, arsenic trioxide (ATO) was shown to rescue the conformation of p53 structural mutants by stabilizing the DNA binding domain. We examined the responses of 17 frequently observed p53 TS mutants to functional rescue by temperature shift and ATO. The results showed that ATO only rescued mild p53 TS mutants with high basal activity at 37°C. Mild TS mutants showed a common feature of regaining significant activity at the semi-permissive temperature of 35°C and could be further stimulated by ATO at 35°C. TS p53 rescue by ATO was antagonized by the cellular redox mechanism and was rapidly reversible. Inhibition of glutathione (GSH) biosynthesis enhanced ATO rescue efficiency and sustained p53 activity after ATO washout. The results suggest that mild TS p53 mutants are uniquely responsive to functional rescue by ATO due to small thermostability deficits and inherent potential to regain active conformation. Combining mild hypothermia and ATO may provide an effective and safe procedure for targeting tumors with p53 TS mutations.
Collapse
Affiliation(s)
- Junhao Lu
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Lihong Chen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Zainab Fatima
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jeffrey Huang
- Department of Anesthesiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jiandong Chen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Allegra A, Mirabile G, Caserta S, Stagno F, Russo S, Pioggia G, Gangemi S. Oxidative Stress and Chronic Myeloid Leukemia: A Balance between ROS-Mediated Pro- and Anti-Apoptotic Effects of Tyrosine Kinase Inhibitors. Antioxidants (Basel) 2024; 13:461. [PMID: 38671909 PMCID: PMC11047441 DOI: 10.3390/antiox13040461] [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: 02/20/2024] [Revised: 03/31/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The balanced reciprocal translocation t (9; 22) (q34; q11) and the BCR-ABL fusion gene, which produce p210 bcr-abl protein production with high tyrosine kinase activity, are characteristics of chronic myeloid leukemia, a myeloproliferative neoplasm. This aberrant protein affects several signaling pathways connected to both apoptosis and cell proliferation. It has been demonstrated that tyrosine kinase inhibitor treatment in chronic myeloid leukemia acts by inducing oxidative stress and, depending on its level, can activate signaling pathways responsible for either apoptosis or survival in leukemic cells. Additionally, oxidative stress and reactive oxygen species generation also mediate apoptosis through genomic activation. Furthermore, it was shown that oxidative stress has a role in both BCR-ABL-independent and BCR-ABL-dependent resistance pathways to tyrosine kinases, while patients with chronic myeloid leukemia were found to have a significantly reduced antioxidant level. The ideal environment for tyrosine kinase inhibitor therapy is produced by a favorable oxidative status. We discuss the latest studies that aim to manipulate the redox system to alter the apoptosis of cancerous cells.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Fabio Stagno
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Sabina Russo
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy;
| |
Collapse
|
4
|
Jiang Y, Shen X, Zhi F, Wen Z, Gao Y, Xu J, Yang B, Bai Y. An overview of arsenic trioxide-involved combined treatment algorithms for leukemia: basic concepts and clinical implications. Cell Death Discov 2023; 9:266. [PMID: 37500645 PMCID: PMC10374529 DOI: 10.1038/s41420-023-01558-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Arsenic trioxide is a first-line treatment drug for acute promyelocytic leukemia, which is also effective for other kinds of leukemia. Its side effects, however, limit its clinical application, especially for patients with complex leukemia symptoms. Combination therapy can effectively alleviate these problems. This review summarizes the research progress on the combination of arsenic trioxide with anticancer drugs, vitamin and vitamin analogs, plant products, and other kinds of drugs in the treatment of leukemia. Additionally, the new progress in arsenic trioxide-induced cardiotoxicity was summarized. This review aims to provide new insights for the rational clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Yanan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| | - Xiuyun Shen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Fengnan Zhi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhengchao Wen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yang Gao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences (2019RU070), Harbin, China.
| | - Yunlong Bai
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
5
|
Sumi D, Taguchi H, Takeuchi K, Fujishiro H. CHAC1 exacerbates arsenite cytotoxicity by lowering intracellular glutathione levels. J Toxicol Sci 2023; 48:487-494. [PMID: 37661365 DOI: 10.2131/jts.48.487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
We here examined whether CHAC1 is implicated in arsenite (As(III))-induced cytotoxicity in HaCaT cells. We found that HaCaT cells in which the intracellular GSH levels were elevated by transfection with CHAC1 siRNA showed decreased sensitivity to As(III) compared to the control cells. Treatment with BSO (an inhibitor of GSH biosynthesis) abolished the decrease in sensitivity to As(III), suggesting that an increase in intracellular GSH levels was involved in the decrease in sensitivity to As(III) due to the decrease in the levels of CHAC1 expression. When we examined the expression of CHAC1 after exposure of HaCaT cells to As(III), the levels of CHAC1 were increased. Since CHAC1 is a proapoptotic factor, we examined appearance of apoptotic cells and cleavage of caspase-3 after exposure to As(III) to determine whether As(III)-induced CHAC1 up-regulation was involved in apoptosis induction. The results showed that induction of apoptosis by As(III) exposure was not detected in CHAC1 siRNA-transfected cells. Together, our findings indicate that CHAC1 is involved in the sensitivity of HaCaT cells to As(III) by regulating the intracellular GSH levels, and in particular, CHAC1 is involved in As(III)-induced apoptosis.
Collapse
Affiliation(s)
- Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Hiroki Taguchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Kumiko Takeuchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | | |
Collapse
|
6
|
Wahiduzzaman M, Ota A, Hosokawa Y. Novel Mechanistic Insights into the Anti-cancer Mode of Arsenic Trioxide. Curr Cancer Drug Targets 2021; 20:115-129. [PMID: 31736446 DOI: 10.2174/1568009619666191021122006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Arsenic, a naturally-occurring toxic element, and a traditionally-used drug, has received a great deal of attention worldwide due to its curative anti-cancer properties in patients with acute promyelocytic leukemia. Among the arsenicals, arsenic trioxide has been most widely used as an anti-cancer drug. Recent advances in cancer therapeutics have led to a paradigm shift away from traditional cytotoxic drugs towards the targeting of proteins closely associated with driving the cancer phenotype. Due to the diverse anti-cancer effects of ATO on different types of malignancies, numerous studies have made efforts to uncover the mechanisms of ATO-induced tumor suppression. From in vitro cellular models to studies in clinical settings, ATO has been extensively studied. The outcomes of these studies have opened doors to establishing improved molecular-targeted therapies for cancer treatment. The efficacy of ATO has been augmented by combination with other drugs. In this review, we discuss recent arsenic-based cancer therapies and summarize the novel underlying molecular mechanisms of the anti-cancer effects of ATO.
Collapse
Affiliation(s)
- Md Wahiduzzaman
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
7
|
Corso CR, Acco A. Glutathione system in animal model of solid tumors: From regulation to therapeutic target. Crit Rev Oncol Hematol 2018; 128:43-57. [DOI: 10.1016/j.critrevonc.2018.05.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/10/2018] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
|
8
|
Wang R, Li Y, Gong P, Gabrilove J, Waxman S, Jing Y. Arsenic Trioxide and Sorafenib Induce Synthetic Lethality of FLT3-ITD Acute Myeloid Leukemia Cells. Mol Cancer Ther 2018; 17:1871-1880. [PMID: 29959200 DOI: 10.1158/1535-7163.mct-17-0298] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 01/05/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) with Fms-related tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation is notoriously hard to treat. We identified two drugs that together form an effective combination therapy against FLT3-ITD AML. One of the drugs, Sorafenib, an inhibitor of FLT3-ITD and other kinase activity, produces an impressive but short-lived remission in FLT3-ITD AML patients. The second, arsenic trioxide (ATO), at therapeutically achievable concentrations, reduces the level of FLT3-ITD and Mcl-1 proteins, and induces apoptosis in leukemic cell lines and in primary cells expressing FLT3-ITD. We linked this relative sensitivity to ATO to low levels of reduced glutathione. While producing proapoptotic effects, ATO treatment also has an unwanted effect whereby it causes the accumulation of the phosphorylated (inactive) form of glycogen synthase kinase 3β (GSK3β), a kinase necessary for apoptosis. When ATO is combined with Sorafenib, GSK3β is activated, Mcl-1 is further reduced, and proapoptotic proteins Bak and Bax are activated. Mice xenografted with FLT3-ITD MOLM13 cell line treated with the Sorafenib/ATO combination have significantly improved survival. This combination has potential to improve the therapeutic outcome of FLT3-ITD-targeted therapy of AML patients. Mol Cancer Ther; 17(9); 1871-80. ©2018 AACR.
Collapse
Affiliation(s)
- Rui Wang
- Division of Hematology/Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Ying Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ping Gong
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Janice Gabrilove
- Division of Hematology/Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Samuel Waxman
- Division of Hematology/Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Yongkui Jing
- Division of Hematology/Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York. .,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
9
|
Organic arsenicals target thioredoxin reductase followed by oxidative stress and mitochondrial dysfunction resulting in apoptosis. Eur J Med Chem 2018; 143:1090-1102. [DOI: 10.1016/j.ejmech.2017.05.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/21/2017] [Accepted: 05/06/2017] [Indexed: 11/23/2022]
|
10
|
Abstract
Purpose of review The goal of this review is to summarize recent advances in our understanding of the regulation of redox homeostasis and the subtype-specific role of antioxidant enzymes in B-cell-derived malignancies. Furthermore, it presents selected prooxidative therapeutic strategies against B-cell neoplasms. Recent findings Recent reports have shown that the disturbed redox homeostasis in B-cell malignancies is regulated by cancer-specific signaling pathways and therefore varies between the individual subtypes. For instance, in a subtype of diffuse large B-cell lymphoma with increased oxidative phosphorylation, elevated reactive oxygen species are accompanied by higher levels of thioredoxin and glutathione and inhibition of either of these systems is selectively toxic to this subtype. In addition, growing number of small molecule inhibitors targeting antioxidant enzymes, such as auranofin, SK053, adenanthin, or decreasing glutathione level, such as imexon, buthionine sulfoximine, and L-cysteinase, trigger specific cytotoxic effects against B-cell malignancies. Lastly, attention is drawn to recent reports of effective treatment modalities involving prooxidative agents and interfering with redox homeostasis provided by stromal cells. Summary Recent findings reveal important differences in redox homeostasis within the distinct subsets of B-cell-derived malignancies that can be therapeutically exploited to improve existing treatment and to overcome drug resistance.
Collapse
|
11
|
Ndombera FT. Anti-cancer agents and reactive oxygen species modulators that target cancer cell metabolism. PURE APPL CHEM 2017. [DOI: 10.1515/pac-2016-1219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AbstractTraditionally the perspective on reactive oxygen species (ROS) has centered on the role they play as carcinogenic or cancer-causing radicals. Over the years, characterization and functional studies have revealed the complexity of ROS as signaling molecules that regulate various physiological cellular responses or whose levels are altered in various diseases. Cancer cells often maintain high basal level of ROS and are vulnerable to any further increase in ROS levels beyond a certain protective threshold. Consequently, ROS-modulation has emerged as an anticancer strategy with synthesis of various ROS-inducing or responsive agents that target cancer cells. Of note, an increased carbohydrate uptake and/or induction of death receptors of cancer cells was exploited to develop glycoconjugates that potentially induce cellular stress, ROS and apoptosis. This mini review highlights the development of compounds that target cancer cells by taking advantage of redox or metabolic alteration in cancer cells.
Collapse
|
12
|
3-bromopyruvate and buthionine sulfoximine effectively kill anoikis-resistant hepatocellular carcinoma cells. PLoS One 2017; 12:e0174271. [PMID: 28362858 PMCID: PMC5376082 DOI: 10.1371/journal.pone.0174271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/06/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND & AIMS Acquisition of anoikis resistance is a prerequisite for metastasis in hepatocellular carcinoma (HCC). However, little is known about how energy metabolism and antioxidant systems are altered in anoikis-resistant (AR) HCC cells. We evaluated anti-tumor effects of a combination treatment of 3-bromopyruvate (3-BP) and buthionine sulfoximine (BSO) in AR HCC cells. METHODS We compared glycolysis, reactive oxygen species (ROS) production, and chemoresistance among Huh-BAT, HepG2 HCC cells, and the corresponding AR cells. Expression of hexokinase II, gamma-glutamylcysteine synthetase (rGCS), and epithelial-mesenchymal transition (EMT) markers in AR cells was assessed. Anti-tumor effects of a combination treatment of 3-BP and BSO were evaluated in AR cells and an HCC xenograft mouse model. RESULTS AR HCC cells showed significantly higher chemoresistance, glycolysis and lower ROS production than attached cells. Expression of hexokinase II, rGCS, and EMT markers was higher in AR HCC cells than attached cells. A combination treatment of 3-BP/BSO effectively suppressed proliferation of AR HCC cells through apoptosis by blocking glycolysis and enhancing ROS levels. In xenograft mouse models, tumor growth derived from AR HCC cells was significantly suppressed in the group treated with 3-BP/BSO compared to the group treated with 3-BP or sorafenib. CONCLUSIONS These results demonstrated that a combination treatment of 3-BP/BSO had a synergistic anti-tumor effect in an AR HCC model. This strategy may be an effective adjuvant therapy for patients with sorafenib-resistant HCC.
Collapse
|
13
|
Ciarcia R, Damiano S, Puzio MV, Montagnaro S, Pagnini F, Pacilio C, Caparrotti G, Bellan C, Garofano T, Polito MS, Giordano A, Florio S. Comparison of Dasatinib, Nilotinib, and Imatinib in the Treatment of Chronic Myeloid Leukemia. J Cell Physiol 2015; 231:680-7. [PMID: 26235483 DOI: 10.1002/jcp.25118] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 07/30/2015] [Indexed: 11/06/2022]
Abstract
To overcome the drug resistance phenomenon induced by Imatibib (IM), in clinical practice, are often used second generation of tyrosine kinase inhibitors as Nilotinib (NIL); a such potent inhibitor of the BCR/ABL kinase and Dasatinib (DAS), a inhibitor of BCR/ABL kinase, and inhibitor SrC family kinase. In this study we evaluated the in vivo effect of DAS, NIL, and IM on intracellular calcium concentration, oxidative stress, and apoptosis in peripheral blood leukocytes of 45 newly diagnosed patients with chronic myeloid leukaemia (CML-PBM). Our data demonstrated that treatment with DAS and NIL showed an higher modulating potential than IM on intracellular calcium concentration by inhibiting the thapsigargin, a sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor, and Lithium (Li) an inositol 1,4,5-triphosphate (InsP3) receptor inhibitor activities. Moreover our data demonstrated that NIL and DAS have significantly increased apoptosis more than IM by involving both intracellular calcium signaling as well as oxidative stress. The acquisition of the oxidative stress and calcium channels receptors values data could help the hematologist to modulate and improve the treatment of chronic myeloid leukaemia (CML) pathology.
Collapse
Affiliation(s)
- Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Maria Valeria Puzio
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Francesco Pagnini
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Carmen Pacilio
- Clinical Trials Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Giuseppe Caparrotti
- Department of Haematology, ASL Caserta (CE), Hospital Moscati, Aversa (CE), Italy
| | - Cristiana Bellan
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Maria Sole Polito
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.,AORN Dei Colli Monaldi UOC, Oncology, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Salvatore Florio
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
14
|
Schoeneberger H, Belz K, Schenk B, Fulda S. Impairment of antioxidant defense via glutathione depletion sensitizes acute lymphoblastic leukemia cells for Smac mimetic-induced cell death. Oncogene 2014; 34:4032-43. [PMID: 25381820 DOI: 10.1038/onc.2014.338] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 08/06/2014] [Accepted: 08/29/2014] [Indexed: 12/18/2022]
Abstract
Evasion of apoptosis in pediatric acute lymphoblastic leukemia (ALL) is linked to aberrant expression of inhibitor of apoptosis (IAP) proteins and dysregulated redox homeostasis, rendering leukemic cells vulnerable to redox-targeting therapies. Here we discover that inhibition of antioxidant defenses via glutathione (GSH) depletion by buthionine sulfoximine (BSO) primes ALL cells for apoptosis induced by the Smac mimetic BV6 that antagonizes IAP proteins. Similarly, BSO cooperates with BV6 to induce cell death in patient-derived primary leukemic samples, underscoring the clinical relevance. In contrast, BSO does not sensitize non-malignant lymphohematopoietic cells from healthy donors toward BV6, pointing to some tumor selectivity. Mechanistically, both agents cooperate to stimulate reactive oxygen species (ROS) production, which is required for BSO/BV6-induced cell death, as ROS inhibitors (that is, N-acetylcysteine, MnTBAP, Trolox) significantly rescue cell death. Further, BSO and BV6 cooperate to trigger lipid peroxidation, which is necessary for cell death, as genetic or pharmacological blockage of lipid peroxidation by GSH peroxidase 4 (GPX4) overexpression or α-tocopherol significantly inhibits BSO/BV6-mediated cell death. Consistently, GPX4 knockdown or GPX4 inhibitor RSL3 enhances lipid peroxidation and cell death by BSO/BV6 cotreatment. The discovery of redox regulation of Smac mimetic-induced cell death has important implications for developing rational Smac mimetic-based combination therapies.
Collapse
Affiliation(s)
- H Schoeneberger
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - K Belz
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - B Schenk
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| |
Collapse
|
15
|
Li X, Liu X, Wang L, Lv X, Li P, Lu K, Wang X. Effects of arsenic sulfide (As 2S 2) on B and T lymphoma cell lines and possible underlying mechanisms. Biomed Rep 2014; 1:583-588. [PMID: 24648990 DOI: 10.3892/br.2013.119] [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: 01/01/2013] [Accepted: 05/25/2013] [Indexed: 11/06/2022] Open
Abstract
Lymphoma is a hematological malignancy that originates from lymph nodes and lymphoid tissues and is divided into Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL), based on its histopathological characteristics. The aim of this study was to investigate the effects of arsenic sulfide (As2S2), the main ingredient of realgar, on the proliferation and apoptosis of the Raji B-cell lymphoma and Jurkat T-cell lymphoma lines, comparing the sensitivity between the two cell lines and investigating the possible underlying mechanisms. The two lymphoma cell lines were cultured in vitro, using different concentrations of As2S2 for different time periods. The cell proliferation was detected using the Cell Counting kit-8 (CCK-8). Apoptosis was assessed via flow cytometry. Expression levels of the apoptosis-associated genes [Homo sapiens Bcl-2-associated X protein (BAX), Homo sapiens B-cell CLL/lymphoma 2 (Bcl-2), Homo sapiens Bcl-2-like protein 1 (BCL2L1, Bcl-xL), Homo sapiens v-myc myelocytomatosis viral oncogene homolog (avian) (MYC, c-Myc) and Homo sapiens pim-1 oncogene (PIM)] were measured via the reverse transcription polymerase chain reaction (RT-PCR) method. The results demonstrated that As2S2 inhibited proliferation and induced apoptosis in the two lymphoma cell lines in a time- and concentration-dependent manner, with the Raji cells being more sensitive to As2S2 compared to Jurkat cells. As2S2 may also alter the expression levels of different apoptosis-associated genes, with the alterations of the mRNA expression levels being different between Raji and Jurkat cells. These findings indicated that As2S2 may inhibit the proliferation and promote the apoptosis of non-Hodgkin lymphoma (NHL) cell lines and that B-cell lymphoma cell lines are more sensitive compared to T-cell lymphoma cell lines. The possible underlying mechanism is that As2S2 alters the expression levels of the apoptosis-associated genes and activates apoptosis-associated signaling pathways.
Collapse
Affiliation(s)
- Xianglu Li
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Xinyu Liu
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Ling Wang
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Xiao Lv
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Peipei Li
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Kang Lu
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, P.R. China
| |
Collapse
|
16
|
Additivity, antagonism, and synergy in arsenic trioxide-induced growth inhibition of C6 glioma cells: effects of genistein, quercetin and buthionine-sulfoximine. Food Chem Toxicol 2014; 67:212-21. [PMID: 24632069 DOI: 10.1016/j.fct.2014.02.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/29/2014] [Accepted: 02/27/2014] [Indexed: 01/09/2023]
Abstract
Arsenic trioxide (ATO) induces clinical remission in acute promyelocytic leukemia and growth inhibition in various cancer cell lines in vitro. Recently, genistein and quercetin were reported to potentiate ATO-provoked apoptosis in leukemia and hepatocellular carcinoma cells. Genistein acted via enhanced ROS generation and quercetin via glutathione depletion. Searching for potential strategies for the treatment of malignant gliomas in this study the capacity of these flavonoids to sensitize rat C6 astroglioma cells for the cytotoxic action of ATO was investigated. ATO inhibited cell growth in a concentration- and time-dependent manner. This effect was accompanied neither by enhanced radical generation nor lipid peroxidation and was not attributed to apoptosis. ATO treatment concentration-dependently increased glutathione levels. Genistein enhanced radical generation. Combined with ATO it inhibited cell growth additively. Additivity was also obtained after cotreatment with ATO and H2O2. Quercetin acted antagonistically on ATO-induced growth inhibition. Quercetin increased glutathione levels. In contrast, buthionine-sulfoximine (BSO) depleted cellular glutathione and acted synergistically with ATO. In conclusion, in C6 cells neither genistein nor quercetin are suited as sensitizing agent, in contrast to BSO. Depletion of cellular glutathione content rather than an increase of ROS generation plays a central role in the enhancement of ATO-toxicity in C6 cells.
Collapse
|
17
|
Tanaka Y, Komatsu T, Shigemi H, Yamauchi T, Fujii Y. BIMEL is a key effector molecule in oxidative stress-mediated apoptosis in acute myeloid leukemia cells when combined with arsenic trioxide and buthionine sulfoximine. BMC Cancer 2014; 14:27. [PMID: 24428916 PMCID: PMC4029189 DOI: 10.1186/1471-2407-14-27] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/10/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Arsenic trioxide (ATO) is reported to be an effective therapeutic agent in acute promyelocytic leukemia (APL) through inducing apoptotic cell death. Buthionine sulfoximine (BSO), an oxidative stress pathway modulator, is suggested as a potential combination therapy for ATO-insensitive leukemia. However, the precise mechanism of BSO-mediated augmentation of ATO-induced apoptosis is not fully understood. In this study we compared the difference in cell death of HL60 leukemia cells treated with ATO/BSO and ATO alone, and investigated the detailed molecular mechanism of BSO-mediated augmentation of ATO-induced cell death. METHODS HL60 APL cells were used for the study. The activation and expression of a series of signal molecules were analyzed with immunoprecipitation and immunoblotting. Apoptotic cell death was detected with caspases and poly (ADP-ribose) polymerase activation. Generation of intracellular reactive oxygen species (ROS) was determined using a redox-sensitive dye. Mitochondrial outer membrane permeabilization was observed with a confocal microscopy using NIR dye and cytochrome c release was determined with immunoblotting. Small interfering (si) RNA was used for inhibition of gene expression. RESULTS HL60 cells became more susceptible to ATO in the presence of BSO. ATO/BSO-induced mitochondrial injury was accompanied by reduced mitochondrial outer membrane permeabilization, cytochrome c release and caspase activation. ATO/BSO-induced mitochondrial injury was inhibited by antioxidants. Addition of BSO induced phosphorylation of the pro-apoptotic BCL2 protein, BIMEL, and anti-apoptotic BCL2 protein, MCL1, in treated cells. Phosphorylated BIMEL was dissociated from MCL1 and interacted with BAX, followed by conformational change of BAX. Furthermore, the knockdown of BIMEL with small interfering RNA inhibited the augmentation of ATO-induced apoptosis by BSO. CONCLUSIONS The enhancing effect of BSO on ATO-induced cell death was characterized at the molecular level for clinical use. Addition of BSO induced mitochondrial injury-mediated apoptosis via the phosphorylation of BIMEL and MCL1, resulting in their dissociation and increased the interaction between BIMEL and BAX.
Collapse
Affiliation(s)
| | - Takayuki Komatsu
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1 Yazako-Karimata, Nagakute, Aichi, Japan.
| | | | | | | |
Collapse
|
18
|
Regulatory Effects of Arsenic on Cellular Signaling Pathways: Biological Effects and Therapeutic Implications. NUCLEAR SIGNALING PATHWAYS AND TARGETING TRANSCRIPTION IN CANCER 2014. [DOI: 10.1007/978-1-4614-8039-6_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
19
|
Synergistic apoptosis of CML cells by buthionine sulfoximine and hydroxychavicol correlates with activation of AIF and GSH-ROS-JNK-ERK-iNOS pathway. PLoS One 2013; 8:e73672. [PMID: 24040019 PMCID: PMC3767640 DOI: 10.1371/journal.pone.0073672] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Hydroxychavicol (HCH), a constituent of Piper betle leaf has been reported to exert anti-leukemic activity through induction of reactive oxygen species (ROS). The aim of the study is to optimize the oxidative stress –induced chronic myeloid leukemic (CML) cell death by combining glutathione synthesis inhibitor, buthionine sulfoximine (BSO) with HCH and studying the underlying mechanism. Materials and Methods Anti-proliferative activity of BSO and HCH alone or in combination against a number of leukemic (K562, KCL22, KU812, U937, Molt4), non-leukemic (A549, MIA-PaCa2, PC-3, HepG2) cancer cell lines and normal cell lines (NIH3T3, Vero) was measured by MTT assay. Apoptotic activity in CML cell line K562 was detected by flow cytometry (FCM) after staining with annexinV-FITC/propidium iodide (PI), detection of reduced mitochondrial membrane potential after staining with JC-1, cleavage of caspase- 3 and poly (ADP)-ribose polymerase proteins by western blot analysis and translocation of apoptosis inducing factor (AIF) by confocal microscopy. Intracellular reduced glutathione (GSH) was measured by colorimetric assay using GSH assay kit. 2′,7′-dichlorodihydrofluorescein diacetate (DCF-DA) and 4-amino-5-methylamino-2′,7′-difluorofluorescein (DAF-FM) were used as probes to measure intracellular increase in ROS and nitric oxide (NO) levels respectively. Multiple techniques like siRNA transfection and pharmacological inhibition were used to understand the mechanisms of action. Results Non-apoptotic concentrations of BSO significantly potentiated HCH-induced apoptosis in K562 cells. BSO potentiated apoptosis-inducing activity of HCH in CML cells by caspase-dependent as well as caspase-independent but apoptosis inducing factor (AIF)-dependent manner. Enhanced depletion of intracellular GSH induced by combined treatment correlated with induction of ROS. Activation of ROS- dependent JNK played a crucial role in ERK1/2 activation which subsequently induced the expression of inducible nitric oxide synthase (iNOS). iNOS- mediated production of NO was identified as an effector molecule causing apoptosis of CML cells. Conclusion/Significance BSO synergizes with HCH in inducing apoptosis of CML cells through the GSH-ROS-JNK-ERK-iNOS pathway.
Collapse
|
20
|
Wildner G, Kaufmann U. What causes relapses of autoimmune diseases? The etiological role of autoreactive T cells. Autoimmun Rev 2013; 12:1070-5. [DOI: 10.1016/j.autrev.2013.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 04/25/2013] [Indexed: 10/26/2022]
|
21
|
Role of glutathione in cancer progression and chemoresistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:972913. [PMID: 23766865 PMCID: PMC3673338 DOI: 10.1155/2013/972913] [Citation(s) in RCA: 762] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/26/2013] [Accepted: 04/29/2013] [Indexed: 01/19/2023]
Abstract
Glutathione (GSH) plays an important role in a multitude of cellular processes, including cell differentiation, proliferation, and apoptosis, and disturbances in GSH homeostasis are involved in the etiology and progression of many human diseases including cancer. While GSH deficiency, or a decrease in the GSH/glutathione disulphide (GSSG) ratio, leads to an increased susceptibility to oxidative stress implicated in the progression of cancer, elevated GSH levels increase the antioxidant capacity and the resistance to oxidative stress as observed in many cancer cells. The present review highlights the role of GSH and related cytoprotective effects in the susceptibility to carcinogenesis and in the sensitivity of tumors to the cytotoxic effects of anticancer agents.
Collapse
|
22
|
Palakurthi S, Yellepeddi VK, Vangara KK. Recent trends in cancer drug resistance reversal strategies using nanoparticles. Expert Opin Drug Deliv 2013; 9:287-301. [PMID: 22339554 DOI: 10.1517/17425247.2012.665365] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Resistance to chemotherapy is a major obstacle in the successful amelioration of tumors in many cancer patients. Resistance is either intrinsic or acquired, involving mechanisms such as genetic aberrations, decreased influx and increased efflux of drugs. Strategies for the reversal of resistance involve the alteration of enzymes responsible for drug resistance, the modulation of proteins regulating apoptosis mechanisms and improving the uptake of drugs using nanotechnology. Novel strides in the reversal of drug resistance are emerging, involving the use of nanotechnology, targeting stem cells, etc. AREAS COVERED This paper reviews the most recent cancer drug reversal strategies involving nanotechnology for targeting cancer cells and cancer stem cells (CSCs), for enhanced uptake of micro- and macromolecular inhibitors. EXPERT OPINION Nanotechnology used in conjunction with existing therapies, such as gene therapy and P-glycoprotein inhibition, has been shown to improve the reversal of drug resistance; the mechanisms involved in this include specific targeting of drugs and nucleotide therapeutics, enhanced cellular uptake of drugs and improved bioavailability of drugs with poor physicochemical characteristics. Important strategies in the reversal of drug resistance include: a multifunctional nanoparticulate system housing a targeting moiety; therapeutics to kill resistant cancer cells and CSCs; cytotoxic drugs and a tumor microenvironment stimuli-responsive element, to release the encapsulated therapeutics.
Collapse
Affiliation(s)
- Srinath Palakurthi
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA.
| | | | | |
Collapse
|
23
|
Wang R, Liu C, Xia L, Zhao G, Gabrilove J, Waxman S, Jing Y. Ethacrynic acid and a derivative enhance apoptosis in arsenic trioxide-treated myeloid leukemia and lymphoma cells: the role of glutathione S-transferase p1-1. Clin Cancer Res 2012; 18:6690-701. [PMID: 23082001 DOI: 10.1158/1078-0432.ccr-12-0770] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Arsenic trioxide (ATO) as a single agent is used for treatment of acute promyelocytic leukemia (APL) with minimal toxicity, but therapeutic effect of ATO in other types of malignancies has not been achieved. We tested whether a combination with ethacrynic acid (EA), a glutathione S-transferase P1-1 (GSTP1-1) inhibitor, and a reactive oxygen species (ROS) inducer will extend the therapeutic effect of ATO beyond APL. EXPERIMENTAL DESIGN The combined apoptotic effects of ATO plus ethacrynic acid were tested in non-APL leukemia and lymphoma cell lines. The role of ROS, GSTP1-1, glutathione (GSH), and Mcl-1 in apoptosis was determined. The selective response to this combination of cells with and without GSTP1-1 expression was compared. RESULTS ATO/EA combination synergistically induced apoptosis in myeloid leukemia and lymphoma cells. This treatment produced high ROS levels, activated c-jun-NH(2)-kinase (JNK), and reduced Mcl-1 protein. This led to the decrease of mitochondrial transmembrane potential, release of cytochrome c, and subsequently, to activation of caspase-3 and -9. Induction of apoptosis in leukemia and lymphoma cells expressing GSTP1-1 required high ethacrynic acid concentrations to be combined with ATO. Silencing of GSTP1 in leukemia cells sensitized them to ATO/EA-induced apoptosis. In a subgroup of B-cell lymphoma, which does not express GSTP1-1, lower concentrations of ethacrynic acid and its more potent derivative, ethacrynic acid butyl-ester (EABE), decreased intracellular GSH levels and synergistically induced apoptosis when combined with ATO. CONCLUSION B-cell lymphoma cells lacking GSTP1-1 are more sensitive than myeloid leukemia cells to ATO/EA-induced apoptosis.
Collapse
Affiliation(s)
- Rui Wang
- The Division of Hematology/Oncology, Department of Medicine, The Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Wang R, Xia L, Gabrilove J, Waxman S, Jing Y. Downregulation of Mcl-1 through GSK-3β activation contributes to arsenic trioxide-induced apoptosis in acute myeloid leukemia cells. Leukemia 2012; 27:315-24. [PMID: 22751450 PMCID: PMC3478411 DOI: 10.1038/leu.2012.180] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Arsenic trioxide (ATO) induces disease remission in acute promyelocytic leukemia (APL) patients, but not in non-APL acute myeloid leukemia (AML) patients. ATO at therapeutic concentrations (1-2 μM) induce APL NB4, but not non-APL HL-60, cells to undergo apoptosis through the mitochondrial pathway. The role of antiapoptotic protein Mcl-1 in ATO-induced apoptosis was determined. The levels of Mcl-1 were decreased in NB4, but not in HL-60, cells after ATO treatment through proteasomal degradation. Both GSK3β inhibitor SB216763 and siRNA blocked ATO-induced Mcl-1 reduction as well as attenuated ATO-induced apoptosis in NB4 cells. Silencing Mcl-1 sensitized HL-60 cells to ATO-induced apoptosis. Both ERK and AKT inhibitors decreased Mcl-1 levels and enhanced ATO-induced apoptosis in HL-60 cells. Sorafenib, a Raf inhibitor, activated GSK3β by inhibiting its phosphorylation, decreased Mcl-1 levels, and decreased intracellular glutathione levels in HL-60 cells. Sorafenib plus ATO augmented ROS production and apoptosis induction in HL-60 cells and in primary AML cells. These results indicate that ATO induces Mcl-1 degradation through activation of GSK3β in APL cells and provide a rationale for utilizing ATO in combination with sorafenib for the treatment of non-APL AML patients.
Collapse
Affiliation(s)
- R Wang
- The Division of Hematology/Oncology, Department of Medicine, Mount Sinai School of Medicine, The Tisch Cancer Institute, New York, NY, USA
| | | | | | | | | |
Collapse
|
25
|
Beauchamp EM, Uren A. A new era for an ancient drug: arsenic trioxide and Hedgehog signaling. VITAMINS AND HORMONES 2012; 88:333-54. [PMID: 22391311 DOI: 10.1016/b978-0-12-394622-5.00015-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Arsenic has been used for ages as a therapeutic agent. Currently, it is an FDA approved drug to treat acute promyelocytic leukemia where it leads to degradation of the PML-RAR fusion protein. It has been shown to have various other targets in cells such as JNK, NFκB, thioredoxin reductase, and MAPK pathways. Most of its effects in cells have been through arsenic's ability to bind to thiol groups in cysteine residues. Recent evidence has shown that arsenic can inhibit the Hedgehog pathway by inhibiting GLI proteins. The proposed mechanism of action is through direct binding. Potential binding sites include the critical cysteine residues in GLI zinc finger domains. The role of the Hedgehog pathway has been implicated in many cancers such as basal cell carcinoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumors. Current Hedgehog pathway inhibitors have been fraught with resistance issues and so arsenic trioxide may provide an alternative therapy when combined with these other inhibitors or after acquired resistance.
Collapse
Affiliation(s)
- Elspeth M Beauchamp
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | | |
Collapse
|
26
|
Calviño E, Estañ MC, Simón GP, Sancho P, Boyano-Adánez MDC, de Blas E, Bréard J, Aller P. Increased apoptotic efficacy of lonidamine plus arsenic trioxide combination in human leukemia cells. Reactive oxygen species generation and defensive protein kinase (MEK/ERK, Akt/mTOR) modulation. Biochem Pharmacol 2011; 82:1619-29. [DOI: 10.1016/j.bcp.2011.08.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 01/13/2023]
|
27
|
Dolloff NG, Mayes PA, Hart LS, Dicker DT, Humphreys R, El-Deiry WS. Off-target lapatinib activity sensitizes colon cancer cells through TRAIL death receptor up-regulation. Sci Transl Med 2011; 3:86ra50. [PMID: 21653830 DOI: 10.1126/scitranslmed.3001384] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lapatinib, a dual HER2/EGFR (human epidermal growth factor receptor 2/epidermal growth factor receptor) inhibitor, is a recently approved targeted therapy for metastatic breast cancer. Because lapatinib enhances the efficacy of the chemotherapeutic agent capecitabine in breast cancer patients, we tested whether lapatinib also enhances the activity of anticancer agents in colorectal cancer. We found that lapatinib improved the proapoptotic effects of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and two TRAIL receptor agonists, the antibodies mapatumumab and lexatumumab. Tumors from mice treated with a combination of lapatinib and TRAIL exhibited more immunostaining for cleaved caspase-8, a marker of the extrinsic cell death pathway, than did tumors from mice treated with lapatinib or TRAIL alone. Furthermore, combination therapy suppressed tumor growth more effectively than either agent alone. Lapatinib up-regulated the proapoptotic TRAIL death receptors DR4 and DR5, leading to more efficient induction of apoptosis in the presence of TRAIL receptor agonists. This activity of lapatinib was independent of EGFR and HER2. The off-target induction of DR5 by lapatinib resulted from activation of the c-Jun amino-terminal kinase (JNK)/c-Jun signaling axis. This activity of lapatinib on TRAIL death receptor expression and signaling may confer therapeutic benefit when increased doses of lapatinib are used in combination with TRAIL receptor-activating agents.
Collapse
Affiliation(s)
- Nathan G Dolloff
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Department of Medicine, Institute for Translational Medicine and Therapeutics, Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
28
|
Xiao G, Tang X, Yao C, Wang C. Potentiation of arsenic trioxide-induced apoptosis by 8-bromo-7-methoxychrysin in human leukemia cells involves depletion of intracellular reduced glutathione. Acta Biochim Biophys Sin (Shanghai) 2011; 43:712-21. [PMID: 21785114 DOI: 10.1093/abbs/gmr065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The novel chrysin analog 8-bromo-7-methoxychrysin (BrMC) has been reported to induce apoptosis of various cancer cell lines. Arsenic trioxide (ATO) treatment induces clinical remission in acute promyelocytic leukemia patients. The combination of ATO with other agents has been shown to improve therapeutic effectiveness in vitro and in vivo. In this report, the mechanism of apoptosis induced by treatment with ATO alone or in combination with BrMC was studied in U937, HL-60, and Jurkat cells. Our results demonstrated that BrMC cooperated with ATO to induce apoptosis in human leukemia cells. This co-treatment caused mitochondrial transmembrane potential dissipation and stimulated the mitochondrial apoptotic pathway, as evidenced by cytochrome c release, down-regulation of X-linked inhibitor of apoptosis (XIAP) and Bcl-XL, and up-regulation of Bax. BrMC alone or in combination with ATO, decreased Akt phosphorylation as well as intracellular reduced glutathione (GSH) content. The thiol antioxidant N-acetylcysteine and exogenous GSH restored GSH content and attenuated apoptosis induced by co-treatment with ATO plus BrMC. In contrast, the non-thiol antioxidant butylated hydroxyanisole and mannitol failed to do so. These findings suggest that GSH depletion explains at least in part the potentiation of ATO-induced apoptosis by BrMC.
Collapse
Affiliation(s)
- Guangfen Xiao
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | | | | | | |
Collapse
|
29
|
Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Sulindac and its metabolites: Sulindac sulfide and sulindac sulfone enhance cytotoxic effects of arsenic trioxide on leukemic cell lines. Toxicol In Vitro 2011; 25:1075-84. [DOI: 10.1016/j.tiv.2011.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 03/10/2011] [Accepted: 04/07/2011] [Indexed: 12/21/2022]
|
30
|
Assessment of the involvement of oxidative stress and Mitogen-Activated Protein Kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines. Med Oncol 2011; 29:1161-72. [DOI: 10.1007/s12032-011-9920-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022]
|
31
|
Yin S, Wang R, Zhou F, Zhang H, Jing Y. Bcl-xL is a dominant antiapoptotic protein that inhibits homoharringtonine-induced apoptosis in leukemia cells. Mol Pharmacol 2011; 79:1072-83. [PMID: 21415308 DOI: 10.1124/mol.110.068528] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Homoharringtonine (HHT) has been reported to be effective in a portion of patients with acute myeloid leukemia (AML) or chronic myeloid leukemia (CML). To investigate its mechanism of action, cell growth inhibition and cytotoxicity of HHT were investigated in three AML cell lines, HL-60, NB4, and U937, and in three CML cell lines, K562, KU812, and KCL22. AML cells were more sensitive than CML cells to HHT-induced cytotoxicity. Using HL-60 cells, it was revealed that HHT decreased the levels of myeloid cell leukemia 1 (Mcl-1), X-linked inhibitor of apoptosis protein (XIAP), survivin, and B-cell lymphoma 2 (Bcl-2)-homology domain 3 (BH3)-only proteins as well as the mitochondrial membrane potential. The levels of Bcl-2, Bcl-2-associated X protein (Bax), and Bcl-2 homologous antagonist/killer (Bak) proteins in HL-60 cells were not changed after HHT treatment. U937, K562, KU812, and KCL22 cells expressed B-cell lymphoma-extra large (Bcl-xL) and were less responsive to HHT-induced apoptosis than HL-60 cells. Silencing Mcl-1 or Bcl-xL, but not XIAP or survivin, enhanced HHT-induced apoptosis in U937 cells. The levels of HHT-induced apoptosis in K562, KCL22, and KU812 cells were inversely correlated with the levels of Bcl-xL but not those of Bcl-2 or Mcl-1. K562 cells expressing high levels of Bcl-xL but no Bcl-2 were less responsive to HHT-induced apoptosis than KCL22 cells that expressed lower levels of Bcl-xL and higher levels of Bcl-2 protein. In K562 cells, knockdown of Bcl-xL, but not of Mcl-1, enhanced HHT-induced apoptosis. Transfection of Bcl-xL into KCL22 cells attenuated HHT-induced apoptosis. These data suggest that Bcl-xL plays a more important role than Bcl-2 and Mcl-1 in protecting against HHT-induced apoptosis.
Collapse
Affiliation(s)
- Shiliang Yin
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | | | | | | | | |
Collapse
|
32
|
Yu SS, Zhao J, Lei SP, Lin XM, Wang LL, Zhao Y. 4-hydroxybenzyl alcohol ameliorates cerebral injury in rats by antioxidant action. Neurochem Res 2010; 36:339-46. [PMID: 21120604 DOI: 10.1007/s11064-010-0335-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2010] [Indexed: 12/22/2022]
Abstract
4-hydroxybenzyl alcohol (4-HBA), one of the phenolic constituents found in many herbal medicinal plants, exhibits beneficial effects in neurological disorders. In the present study, we evaluated 4-HBA's role in transient cerebral ischemia and its potential mechanism. Pre-treatment with 4-HBA (50,100 mg/kg) significantly reduced the cerebral infarct size and improved the neurological symptoms. Morphological examinations showed 4-HBA reduced the number of degenerated neurons. Oxidative stress was evaluated superoxide dismutase (SOD) activity and malondialdehyde (MDA) level. Anti-oxidative mechanisms were studied by Immunofluorescence staining and western immunoblot analysis. 4-HBA increased the expression of NAD(P)H: quinone oxidoreductase1 (NQO1) and ultimately inhibited oxidative stress. In addition, we evaluated the time course expression of NQO1, which was upregulated in the ischemic brain beginning at 1 h. Taken together, these results suggested that 4-HBA ameliorated cerebral injury in rats, This neuroprotective effect is likely related to its antioxidant activities.
Collapse
Affiliation(s)
- Shan-Shan Yu
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
33
|
Arsenic trioxide as an anti-tumour agent: mechanisms of action and strategies of sensitization. J Appl Biomed 2010. [DOI: 10.2478/v10136-009-0027-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
34
|
Foti C, Florean C, Pezzutto A, Roncaglia P, Tomasella A, Gustincich S, Brancolini C. Characterization of caspase-dependent and caspase-independent deaths in glioblastoma cells treated with inhibitors of the ubiquitin-proteasome system. Mol Cancer Ther 2009; 8:3140-50. [PMID: 19887551 DOI: 10.1158/1535-7163.mct-09-0431] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The regulation of the necrotic death and its relevance in anticancer therapy are largely unknown. Here, we have investigated the proapoptotic and pronecrotic activities of two ubiquitin-proteasome system inhibitors: bortezomib and G5. The present study points out that the glioblastoma cell lines U87MG and T98G are useful models to study the susceptibility to apoptosis and necrosis in response to ubiquitin-proteasome system inhibitors. U87MG cells show resistance to apoptosis induced by bortezomib and G5, but they are more susceptible to necrosis induced by G5. Conversely, T98G cells are more susceptible to apoptosis induced by both inhibitors but show some resistance to G5-induced necrosis. No overt differences in the induction of Noxa and Mcl-1 or in the expression levels of other components of the apoptotic machinery were observed between U87MG and T98G cells. Instead, by comparing the transcriptional profiles of the two cell lines, we have found that the resistance to G5-induced necrosis could arise from differences in glutathione synthesis/utilization and in the microenvironment. In particular, collagen IV, which is highly expressed in T98G cells, and fibronectin, whose adhesive function is counteracted by tenascin-C in U87MG cells, can restrain the necrotic response to G5. Collectively, our results provide an initial characterization of the molecular signals governing cell death by necrosis in glioblastoma cell lines.
Collapse
Affiliation(s)
- Carmela Foti
- Dipartimento di Scienze e Tecnologie Biomediche, Sezione di Biologia Universita' di Udine, Udine, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Sánchez Y, Calle C, de Blas E, Aller P. Modulation of arsenic trioxide-induced apoptosis by genistein and functionally related agents in U937 human leukaemia cells. Regulation by ROS and mitogen-activated protein kinases. Chem Biol Interact 2009; 182:37-44. [DOI: 10.1016/j.cbi.2009.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 08/21/2009] [Accepted: 08/25/2009] [Indexed: 01/04/2023]
|
36
|
Morales AA, Gutman D, Cejas PJ, Lee KP, Boise LH. Reactive oxygen species are not required for an arsenic trioxide-induced antioxidant response or apoptosis. J Biol Chem 2009; 284:12886-95. [PMID: 19279006 DOI: 10.1074/jbc.m806546200] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Arsenicals are both environmental carcinogens as well as therapeutic agents for the treatment of trypanosomiasis and more recently cancer. Arsenic trioxide (ATO) has been successfully used for the treatment of acute promyelocytic leukemia (APL) and has activity in multiple myeloma (MM). While signaling events associated with carcinogenesis have been well studied, it still remains to be determined which of these events are involved in anti-cancer signaling. To better define this response, gene expression profiling following ATO treatment of four MM cell lines was performed. The pattern was consistent with a strong antioxidative response, particularly of genes activated by Nrf2. While Nrf2 is expressed constitutively at the mRNA level, the protein is not detected in untreated cells. Consistent with inactivation of Keap1, Nrf2 protein is stabilized and present in the nucleus within 6 h of ATO treatment. Despite the activation of this antioxidative response, ROS may not be important in ATO-induced death. Inhibition of ATO-induced ROS with butylated hydroxyanisole (BHA) does not affect Nrf2 activation or cell death. Moreover, silencing Nrf2 had no effect on ATO-induced apoptosis. Together these data suggest that ROS is not important in the induction of the antioxidative response or cellular death by ATO.
Collapse
Affiliation(s)
- Alejo A Morales
- Department of Microbiology and Immunology and The Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
37
|
Babu CK, Suwansrinon K, Bren GD, Badley AD, Rizza SA. HIV induces TRAIL sensitivity in hepatocytes. PLoS One 2009; 4:e4623. [PMID: 19247452 PMCID: PMC2644790 DOI: 10.1371/journal.pone.0004623] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 01/23/2009] [Indexed: 02/07/2023] Open
Abstract
Background HIV infected patients have an increased susceptibility to liver disease due to Hepatitis B Virus (HBV), Hepatitis C Virus (HCV), alcoholic, and non-alcoholic steatohepatitis. Clinically, this results in limited options for antiretroviral therapy and accelerated rates of liver disease, causing liver disease to be the second leading cause of death for HIV infected patients. The mechanisms causing this propensity for liver dysfunction during HIV remains unknown. Methodology/Principal Findings We demonstrate that HIV and/or the HIV glycoprotein gp120 ligation of CXCR4 on hepatocytes selectively up-regulates TRAIL R2 expression and confers an acquired sensitivity to TRAIL mediated apoptosis which is mediated by JNK II, but not p38 nor G-proteins. Conclusions/Significance These findings suggest that HIV infection renders hepatocytes more susceptible to liver injury during disease states associated with enhanced TRAIL production such as HBV, HCV, or steatohepatitis.
Collapse
Affiliation(s)
- Challagundla K. Babu
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kanitta Suwansrinon
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gary D. Bren
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Andrew D. Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stacey A. Rizza
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
38
|
Ogino T, Ozaki M, Hosako M, Omori M, Okada S, Matsukawa A. Activation of c-Jun N-terminal kinase is essential for oxidative stress-induced Jurkat cell apoptosis by monochloramine. Leuk Res 2009; 33:151-8. [DOI: 10.1016/j.leukres.2008.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2008] [Revised: 06/26/2008] [Accepted: 07/10/2008] [Indexed: 01/31/2023]
|
39
|
Franco R, Sánchez-Olea R, Reyes-Reyes EM, Panayiotidis MI. Environmental toxicity, oxidative stress and apoptosis: ménage à trois. Mutat Res 2008; 674:3-22. [PMID: 19114126 DOI: 10.1016/j.mrgentox.2008.11.012] [Citation(s) in RCA: 352] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 11/27/2008] [Indexed: 12/21/2022]
Abstract
Apoptosis is an evolutionary conserved homeostatic process involved in distinct physiological processes including organ and tissue morphogenesis, development and senescence. Its deregulation is also known to participate in the etiology of several human diseases including cancer, neurodegenerative and autoimmune disorders. Environmental stressors (cytotoxic agents, pollutants or toxicants) are well known to induce apoptotic cell death and to contribute to a variety of pathological conditions. Oxidative stress seems to be the central element in the regulation of the apoptotic pathways triggered by environmental stressors. In this work, we review the established mechanisms by which oxidative stress and environmental stressors regulate the apoptotic machinery with the aim to underscore the relevance of apoptosis as a component in environmental toxicity and human disease progression.
Collapse
Affiliation(s)
- Rodrigo Franco
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, P. O. Box 12233, 111. T.W. Alexander Drive, Research Triangle Park, NC 27709, United States.
| | | | | | | |
Collapse
|
40
|
Lewis-Wambi JS, Kim HR, Wambi C, Patel R, Pyle JR, Klein-Szanto AJ, Jordan VC. Buthionine sulfoximine sensitizes antihormone-resistant human breast cancer cells to estrogen-induced apoptosis. Breast Cancer Res 2008; 10:R104. [PMID: 19061505 PMCID: PMC2656901 DOI: 10.1186/bcr2208] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 11/11/2008] [Accepted: 12/05/2008] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Estrogen deprivation using aromatase inhibitors is one of the standard treatments for postmenopausal women with estrogen receptor (ER)-positive breast cancer. However, one of the consequences of prolonged estrogen suppression is acquired drug resistance. Our group is interested in studying antihormone resistance and has previously reported the development of an estrogen deprived human breast cancer cell line, MCF-7:5C, which undergoes apoptosis in the presence of estradiol. In contrast, another estrogen deprived cell line, MCF-7:2A, appears to have elevated levels of glutathione (GSH) and is resistant to estradiol-induced apoptosis. In the present study, we evaluated whether buthionine sulfoximine (BSO), a potent inhibitor of glutathione (GSH) synthesis, is capable of sensitizing antihormone resistant MCF-7:2A cells to estradiol-induced apoptosis. METHODS Estrogen deprived MCF-7:2A cells were treated with 1 nM 17beta-estradiol (E2), 100 microM BSO, or 1 nM E2 + 100 microM BSO combination in vitro, and the effects of these agents on cell growth and apoptosis were evaluated by DNA quantitation assay and annexin V and terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) staining. The in vitro results of the MCF-7:2A cell line were further confirmed in vivo in a mouse xenograft model. RESULTS Exposure of MCF-7:2A cells to 1 nM E2 plus 100 microM BSO combination for 48 to 96 h produced a sevenfold increase in apoptosis whereas the individual treatments had no significant effect on growth. Induction of apoptosis by the combination treatment of E2 plus BSO was evidenced by changes in Bcl-2 and Bax expression. The combination treatment also markedly increased phosphorylated c-Jun N-terminal kinase (JNK) levels in MCF-7:2A cells and blockade of the JNK pathway attenuated the apoptotic effect of E2 plus BSO. Our in vitro findings corroborated in vivo data from a mouse xenograft model in which daily administration of BSO either as a single agent or in combination with E2 significantly reduced tumor growth of MCF-7:2A cells. CONCLUSIONS Our data indicates that GSH participates in retarding apoptosis in antihormone-resistant human breast cancer cells and that depletion of this molecule by BSO may be critical in predisposing resistant cells to E2-induced apoptotic cell death. We suggest that these data may form the basis of improving therapeutic strategies for the treatment of antihormone resistant ER-positive breast cancer.
Collapse
Affiliation(s)
- Joan S Lewis-Wambi
- Department of Medical Sciences, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Blatt NB, Boitano AE, Lyssiotis CA, Opipari AW, Glick GD. Bz-423 superoxide signals apoptosis via selective activation of JNK, Bak, and Bax. Free Radic Biol Med 2008; 45:1232-42. [PMID: 18718527 PMCID: PMC2837238 DOI: 10.1016/j.freeradbiomed.2008.07.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Revised: 07/14/2008] [Accepted: 07/16/2008] [Indexed: 02/07/2023]
Abstract
Bz-423 is a proapoptotic 1,4-benzodiazepine with potent therapeutic properties in murine models of lupus and psoriasis. Bz-423 modulates the F(1)F(0)-ATPase, inducing the formation of superoxide within the mitochondrial respiratory chain, which then functions as a second messenger initiating apoptosis. Herein, we report the signaling pathway activated by Bz-423 in mouse embryonic fibroblasts containing knockouts of key apoptotic proteins. Bz-423-induced superoxide activates cytosolic ASK1 and its release from thioredoxin. A mitogen-activated protein kinase cascade follows, leading to the specific phosphorylation of JNK. JNK signals activation of Bax and Bak which then induces mitochondrial outer membrane permeabilization to cause the release of cytochrome c and a commitment to apoptosis. The response of these cells to Bz-423 is critically dependent on both superoxide and JNK activation as antioxidants and the JNK inhibitor SP600125 prevents Bax translocation, cytochrome c release, and cell death. These results demonstrate that superoxide generated from the mitochondrial respiratory chain as a consequence of a respiratory transition can signal a sequential and specific apoptotic response. Collectively, these data suggest that the selectivity of Bz-423 observed in vivo results from cell-type specific differences in redox balance and signaling by ASK1 and Bcl-2 proteins.
Collapse
Affiliation(s)
- Neal B. Blatt
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109
| | - Anthony E. Boitano
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Anthony W. Opipari
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109
| | - Gary D. Glick
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
42
|
Jin HO, Seo SK, Woo SH, Lee HC, Kim ES, Yoo DH, Lee SJ, An S, Choe TB, Kim JI, Hong SI, Rhee CH, Park IC. A combination of sulindac and arsenic trioxide synergistically induces apoptosis in human lung cancer H1299 cells via c-Jun NH2-terminal kinase-dependent Bcl-xL phosphorylation. Lung Cancer 2008; 61:317-27. [DOI: 10.1016/j.lungcan.2008.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 01/01/2008] [Indexed: 12/23/2022]
|
43
|
Sánchez Y, Amrán D, Fernández C, de Blas E, Aller P. Genistein selectively potentiates arsenic trioxide‐induced apoptosis in human leukemia cellsviareactive oxygen species generation and activation of reactive oxygen species‐inducible protein kinases (p38‐MAPK, AMPK). Int J Cancer 2008; 123:1205-14. [DOI: 10.1002/ijc.23639] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
Induction of apoptosis by furanodiene in HL60 leukemia cells through activation of TNFR1 signaling pathway. Cancer Lett 2008; 271:158-66. [PMID: 18667267 DOI: 10.1016/j.canlet.2008.06.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 03/10/2008] [Accepted: 06/02/2008] [Indexed: 11/20/2022]
Abstract
Furanodiene, a natural product isolated from Curcuma wenyujin, has been reported to produce cytotoxic effect. In this study, we investigated its effects on human leukemia HL60 cells. Furanodiene induced apoptosis of HL60 cells, characterized by DNA fragmentation, cleavage of poly (ADP-ribose) polymerase (PARP), caspase-3, caspase-8 and caspase-9. In the Bcl-2 family proteins, Bid protein (a substrate of caspase-8) was activated by furanodiene, but Bcl-2, Bax and Bcl-xL proteins were not influenced by furanodiene stimulation. Moreover, furanodiene treatment caused the upregulation of tumor necrosis factor receptor 1 (TNFR1), the formation of TNFR1 complex and an obvious production of TNF-alpha in HL60 cells. The soluble TNFR1 receptor effectively inhibited furanodiene-induced apoptosis. Taken together, furanodiene could inhibit the growth of leukemia cells via induction of apoptosis, and TNFR1-mediated extrinsic apoptotic pathways explains furanodiene-induced apoptosis.
Collapse
|
45
|
Korashy HM, El-Kadi AOS. NF-κB and AP-1 are key signaling pathways in the modulation of NAD(P)H:Quinone oxidoreductase 1 gene by mercury, lead, and copper. J Biochem Mol Toxicol 2008; 22:274-83. [DOI: 10.1002/jbt.20238] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
46
|
Hosono T, Hosono-Fukao T, Inada K, Tanaka R, Yamada H, Iitsuka Y, Seki T, Hasegawa I, Ariga T. Alkenyl group is responsible for the disruption of microtubule network formation in human colon cancer cell line HT-29 cells. Carcinogenesis 2008; 29:1400-6. [PMID: 18515280 PMCID: PMC2500214 DOI: 10.1093/carcin/bgn124] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alk(en)yl trisulfides (R-SSS-R′) are organosulfur compounds produced by crushed garlic and other Allium vegetables. We found that these compounds exhibit potent anticancer effects through the reaction with microtubules, causing cell cycle arrest. Nine alk(en)yl trisulfides including dimethyl trisulfide, diethyl trisulfide, dipropyl trisulfide (DPTS), dibutyl trisulfide, dipentyl trisulfide, diallyl trisulfide (DATS), dibutenyl trisulfide, dipentenyl trisulfide and allyl methyl trisulfide were synthesized and added to cultures of HT-29 human colon cancer cells at a concentration of 10 μM. The trisulfides with alkenyl groups such as DATS, but not those with alkyl groups, induced rapid microtubule disassembly at 30–60 min as well as cell cycle arrest during the mitotic phase approximately at 4 h after the treatment. Both DATS-induced microtubule disassembly and the cell cycle arrest were cancelled by the simultaneous treatment of the cancer cells with 2 mM L-cysteine, glutathione (GSH) or N-acetyl-L-cysteine. Reciprocally, L-buthionine-(S,R)-sulfoximine (500 μM), an inhibitor of GSH synthesis, enhanced the power of DATS in inducing the cell cycle arrest. These results indicate that alk(en)yl trisulfide react with sulfhydryl groups in cysteine residues of cellular proteins such as microtubule proteins. Thus, the present study provides evidence that trisulfides with alkenyl groups have potent anticancer activities, at least in part, directed toward microtubules. These findings suggest that alkenyl trisulfides and their structurally related compounds may provide novel and effective anticancer agents.
Collapse
Affiliation(s)
- Takashi Hosono
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kameino 1866, Fujisawa, Kanagawa 252-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ramos AM, Aller P. Quercetin decreases intracellular GSH content and potentiates the apoptotic action of the antileukemic drug arsenic trioxide in human leukemia cell lines. Biochem Pharmacol 2008; 75:1912-23. [DOI: 10.1016/j.bcp.2008.02.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 01/30/2008] [Accepted: 02/11/2008] [Indexed: 10/22/2022]
|
48
|
Vernhet L, Morzadec C, van Grevenynghe J, Bareau B, Corolleur M, Fest T, Fardel O. Inorganic arsenic induces necrosis of human CD34-positive haematopoietic stem cells. ENVIRONMENTAL TOXICOLOGY 2008; 23:263-268. [PMID: 18214907 DOI: 10.1002/tox.20334] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Inorganic arsenic is a major environmental contaminant known to exert immunosuppressive effects. In this study, we report toxicity of As2O3, a trivalent inorganic form, toward isolated human hematopoietic CD34+ progenitor cells. Our results demonstrate that low concentrations of As2O3 (0.1-5 microM) inhibit in vitro proliferation of CD34+ cells and their differentiation into various hematological cell lineages. These effects were associated with the induction of a necrotic process independent of caspases and likely related to mitochondrial damage. We conclude that As2O3 can impair in vitro human hematopoiesis by decreasing survival of CD34+ progenitor cells.
Collapse
Affiliation(s)
- Laurent Vernhet
- INSERM U620, Détoxication et Réparation Tissulaire, Université de Rennes 1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France.
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhuo W, Wang Y, Zhuo X, Zhang Y, Ao X, Chen Z. Knockdown of Snail, a novel zinc finger transcription factor, via RNA interference increases A549 cell sensitivity to cisplatin via JNK/mitochondrial pathway. Lung Cancer 2008; 62:8-14. [PMID: 18372076 DOI: 10.1016/j.lungcan.2008.02.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 02/05/2008] [Accepted: 02/14/2008] [Indexed: 12/13/2022]
Abstract
Previous reports have implicated epithelial-mesenchymal transition (EMT) as a major cause of cancer. Snail, a novel zinc finger transcription factor, was suggested to be an important inducer of EMT and therefore be involved in different phases of tumorigenicity. However, whether Snail could increase chemoresistance of cancer cells to chemotherapeutic agent remains unclear. To evaluate the roles and possible mechanisms of Snail in chemoresistance of lung cancer cells to cisplatin, we utilized RNA interference to knockdown Snail expression in A549 cells and further assessed the cell viability and apoptosis as well as possible signaling transduction pathways. The data showed that Snail depletion sensitized A549 cells to cisplatin possibly by inducing activation of JNK/mitochondrial pathway, suggesting critical roles of Snail in A549 cell chemoresistance to cisplatin and raising the possibility of Snail depletion as a promising approach to lung cancer therapy.
Collapse
Affiliation(s)
- Wenlei Zhuo
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
50
|
Korashy HM, El-Kadi AOS. The role of redox-sensitive transcription factors NF-kappaB and AP-1 in the modulation of the Cyp1a1 gene by mercury, lead, and copper. Free Radic Biol Med 2008; 44:795-806. [PMID: 18078826 DOI: 10.1016/j.freeradbiomed.2007.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 11/02/2007] [Accepted: 11/05/2007] [Indexed: 02/06/2023]
Abstract
We have previously shown that Hg(2+), Pb(2+), and Cu(2+) significantly induced the expression of Cyp1a1 mRNA, but the catalytic activity was inhibited by the three metals, and the inhibition was accompanied by an increase in the oxidative stress status. In the current study we investigated the role of redox-sensitive transcription factors and the NF-kappaB and AP-1 signaling pathways in the metal-mediated effects on Cyp1a1 gene expression. We show that heavy metals caused the induction of oxidative stress markers, such as reactive oxygen species and heme oxygenase-1, and the depletion of cellular glutathione content, which was associated with NF-kappaB and AP-1 activation. In addition, the NF-kappaB activator PMA significantly abolished the metal-mediated induction of Cyp1a1 mRNA, whereas it further potentiated their inhibitory effects on Cyp1a1 activity. In parallel, the NF-kappaB inhibitor PDTC further potentiated the metal-mediated induction of Cyp1a1 mRNA, whereas it reversed their inhibitory effects on Cyp1a1 activity. Inhibition of AP-1 upstream signaling pathway activators such as JNK by SP600125 suppressed Cyp1a1 mRNA induction by heavy metals, whereas it potentiated their inhibitory effects at the activity level. In contrast, the ERK inhibitor U0126 further potentiated heavy metal-mediated induction of Cyp1a1 mRNA, whereas it reversed their inhibitory effects on the Cyp1a1 activity. The p38 MAPK inhibitor SB203580 suppressed the metal-mediated induction of Cyp1a1 mRNA, but did not alter Cyp1a1 activity. These results clearly demonstrate that activation of the NF-kappaB and AP-1 signaling pathways is directly involved in the modulation of Cyp1a1 by heavy metals.
Collapse
Affiliation(s)
- Hesham M Korashy
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|