1
|
Tian Z, Jiang S, Zhou J, Zhang W. Copper homeostasis and cuproptosis in mitochondria. Life Sci 2023; 334:122223. [PMID: 38084674 DOI: 10.1016/j.lfs.2023.122223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/30/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023]
Abstract
Mitochondria serve as sites for energy production and are essential for regulating various forms of cell death induced by metal metabolism, targeted anticancer drugs, radiotherapy and immunotherapy. Cuproptosis is an autonomous form of cell death that depends on copper (Cu) and mitochondrial metabolism. Although the recent discovery of cuproptosis highlights the significance of Cu and mitochondria, there is still a lack of biological evidence and experimental verification for the underlying mechanism. We provide an overview of how Cu and cuproptosis affect mitochondrial morphology and function. Through comparison with ferroptosis, similarities and differences in mitochondrial metabolism between cuproptosis and ferroptosis have been identified. These findings provide implications for further exploration of cuproptotic mechanisms. Furthermore, we explore the correlation between cuproptosis and immunotherapy or radiosensitivity. Ultimately, we emphasize the therapeutic potential of targeting cuproptosis as a novel approach for disease treatment.
Collapse
Affiliation(s)
- Ziying Tian
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Su Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Jieyu Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Chen F, Wang L, Feng Y, Ma W, Liu J, Bi Q, Song Y, Gao R, Jia Y. F-box and leucine-rich repeat protein 16 controls tamoxifen sensitivity via regulation of mitochondrial respiration in estrogen receptor-positive breast cancer cells. Hum Cell 2023; 36:2087-2098. [PMID: 37537406 DOI: 10.1007/s13577-023-00961-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
Tamoxifen is one of the most effective therapeutic tools for estrogen receptor-positive (ER +) breast cancer. However, the intrinsic insensitivity and resistance to tamoxifen remains a significant hurdle for achieving optimal responses and curative therapy. In this study, we report that F-box and leucine-rich repeat protein 16 (FBXL16) is located in the mitochondria of ER + breast cancer cells. The mitochondrial FBXL16 plays an essential role in sustaining mitochondrial respiration and thereby regulates the sensitivity of ER + breast cancer cells to tamoxifen treatment. Importantly, high FBXL16 expression is significantly correlated with poor overall survival of ER + breast cancer patients. Moreover, mitochondrial inhibition phenocopies FBXL16 depletion in terms of sensitizing the ER + breast cancer cells to tamoxifen treatment. Together, our study demonstrates that FBXL16 acts as a novel regulator of tamoxifen sensitivity. Thus, targeting FBXL16 may serve as a promising approach for improving the therapeutic efficacy of tamoxifen in ER + breast cancer cells.
Collapse
Affiliation(s)
- Feng Chen
- Department of Medical Oncology, Ordos Central Hospital, Ordos, China
| | - Lu Wang
- Department of Radiation Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanrong Feng
- Ordos School of Clinical Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Wenxin Ma
- Ordos School of Clinical Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Junqin Liu
- Ordos School of Clinical Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Qianyao Bi
- Ordos School of Clinical Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Yao Song
- Department of Radiation Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Gao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China.
| | - Yanhan Jia
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
3
|
Mukherjee AG, Gopalakrishnan AV. The mechanistic insights of the antioxidant Keap1-Nrf2 pathway in oncogenesis: a deadly scenario. Med Oncol 2023; 40:248. [PMID: 37480500 DOI: 10.1007/s12032-023-02124-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/24/2023]
Abstract
The Nuclear factor erythroid 2-related factor 2 (Nrf2) protein has garnered significant interest due to its crucial function in safeguarding cells and tissues. The Nrf2 protein is crucial in preserving tissue integrity by safeguarding cells against metabolic, xenobiotic and oxidative stress. Due to its various functions, Nrf2 is a potential pharmacological target for reducing the incidence of diseases such as cancer. However, mutations in Keap1-Nrf2 are not consistently favored in all types of cancer. Instead, they seem to interact with specific driver mutations of tumors and their respective tissue origins. The Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 pathway mutations are a powerful cancer adaptation that utilizes inherent cytoprotective pathways, encompassing nutrient metabolism and ROS regulation. The augmentation of Nrf2 activity elicits significant alterations in the characteristics of neoplastic cells, such as resistance to radiotherapy and chemotherapy, safeguarding against apoptosis, heightened invasiveness, hindered senescence, impaired autophagy and increased angiogenesis. The altered activity of Nrf2 can arise from diverse genetic and epigenetic modifications that instantly impact Nrf2 regulation. The present study aims to showcase the correlation between the Keap1-Nrf2 pathway and the progression of cancers, emphasizing genetic mutations, metabolic processes, immune regulation, and potential therapeutic strategies. This article delves into the intricacies of Nrf2 pathway anomalies in cancer, the potential ramifications of uncontrolled Nrf2 activity, and therapeutic interventions to modulate the Keap1-Nrf2 pathway.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
Davezac M, Meneur C, Buscato M, Zahreddine R, Arnal JF, Dalenc F, Lenfant F, Fontaine C. The beneficial effects of tamoxifen on arteries: a key player for cardiovascular health of breast cancer patient. Biochem Pharmacol 2023:115677. [PMID: 37419371 DOI: 10.1016/j.bcp.2023.115677] [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: 04/14/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
Breast cancer is the most common cancer in women. Over the past few decades, advances in cancer detection and treatment have significantly improved survival rate of breast cancer patients. However, due to the cardiovascular toxicity of cancer treatments (chemotherapy, anti-HER2 antibodies and radiotherapy), cardiovascular diseases (CVD) have become an increasingly important cause of long-term morbidity and mortality in breast cancer survivors. Endocrine therapies are prescribed to reduce the risk of recurrence and specific death in estrogen receptor-positive (ER+) early breast cancer patients, but their impact on CVD is a matter of debate. Whereas aromatase inhibitors and luteinizing hormone-releasing hormone (LHRH) analogs inhibit estrogen synthesis, tamoxifen acts as a selective estrogen receptor modulator (SERM), opposing estrogen action in the breast but mimicking their actions in other tissues, including arteries. This review aims to summarize the main clinical and experimental studies reporting the effects of tamoxifen on CVD. In addition, we will discuss how recent findings on the mechanisms of action of these therapies may contribute to a better understanding and anticipation of CVD risk in breast cancer patients.
Collapse
Affiliation(s)
- Morgane Davezac
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Cecile Meneur
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France; PhysioStim, 10 rue Henri Regnault, 81100, Castres, France
| | - Melissa Buscato
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Rana Zahreddine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France; CREFRE-Anexplo, Service de Microchirurgie Experimentale, UMS006, INSERM, Université de Toulouse, UT3, ENVT, 31062 Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Florence Dalenc
- Department of Medical Oncology, Claudius Regaud Institute, IUCT-Oncopole, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France.
| |
Collapse
|
5
|
Hwang S, Park S, Kim JH, Bang SB, Kim HJ, Ka NL, Ko Y, Kim SS, Lim GY, Lee S, Shin YK, Park SY, Kim S, Lee MO. Targeting HMG-CoA synthase 2 suppresses tamoxifen-resistant breast cancer growth by augmenting mitochondrial oxidative stress-mediated cell death. Life Sci 2023:121827. [PMID: 37276910 DOI: 10.1016/j.lfs.2023.121827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/07/2023]
Abstract
AIMS In this study, we aimed to investigate previously unrecognized lipid metabolic perturbations in tamoxifen-resistant breast cancer (BC) by conducting comprehensive metabolomics and transcriptomics analysis. We identified the role of 3-hydroxy-3-methylglutary-coenzyme-A-synthase 2 (HMGCS2), a key enzyme responsible for ketogenesis, in tamoxifen-resistant BC growth. MAIN METHODS Comprehensive metabolomics (CE-TOFMS, LC-TOFMS) and transcriptiomics analysis were performed to characterize metabolic pathways in tamoxifen-resistant BC cells. The upregulation of HMGCS2 were verified thorugh immunohistochemistry (IHC) in clinical samples obtained from patients with recurrent BC. HMGCS2 inhibitor was discovered through surface plasmon resonance analysis, enzyme assay, and additional molecular docking studies. The effect of HMGCS2 suppression on tumor growth was studied thorugh BC xenograft model, and intratumoral lipid metabolites were analyzed via MALDI-TOFMS imaging. KEY FINDINGS We revealed that the level of HMGCS2 was highly elevated in both tamoxifen-resistant T47D sublines (T47D/TR) and clinical refractory tumor specimens from patients with ER+ breast cancer, who had been treated with adjuvant tamoxifen. Suppression of HMGCS2 in T47D/TR resulted in the accumulation of mitochondrial reactive oxygen species (mtROS) and apoptotic cell death. Further, we identified alphitolic acid, a triterpenoid natural product, as a novel HMGCS2-specific inhibitor that elevated mtROS levels and drastically retarded the growth of T47D/TR in in vitro and in vivo experiments. SIGNIFICANCE Enhanced ketogenesis with upregulation of HMGCS2 is a potential metabolic vulnerability of tamoxifen-resistant BC that offers a new therapeutic opportunity for treating patients with ER+ BC that are refractory to tamoxifen treatment.
Collapse
Affiliation(s)
- Sewon Hwang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Soojun Park
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jee Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, Republic of Korea
| | - Sang-Beom Bang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Na-Lee Ka
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoonae Ko
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung-Su Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ga Young Lim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seunghee Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, Republic of Korea.
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
6
|
Sfogliarini C, Pepe G, Dolce A, Della Torre S, Cesta MC, Allegretti M, Locati M, Vegeto E. Tamoxifen Twists Again: On and Off-Targets in Macrophages and Infections. Front Pharmacol 2022; 13:879020. [PMID: 35431927 PMCID: PMC9006819 DOI: 10.3389/fphar.2022.879020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022] Open
Abstract
Beyond the wide use of tamoxifen in breast cancer chemotherapy due to its estrogen receptor antagonist activity, this drug is being assayed in repurposing strategies against a number of microbial infections. We conducted a literature search on the evidence related with tamoxifen activity in macrophages, since these immune cells participate as a first line-defense against pathogen invasion. Consistent data indicate the existence of estrogen receptor-independent targets of tamoxifen in macrophages that include lipid mediators and signaling pathways, such as NRF2 and caspase-1, which allow these cells to undergo phenotypic adaptation and potentiate the inflammatory response, without the induction of cell death. Thus, these lines of evidence suggest that the widespread antimicrobial activity of this drug can be ascribed, at least in part, to the potentiation of the host innate immunity. This widens our understanding of the pharmacological activity of tamoxifen with relevant therapeutic implications for infections and other clinical indications that may benefit from the immunomodulatory effects of this drug.
Collapse
Affiliation(s)
- Chiara Sfogliarini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Giovanna Pepe
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Arianna Dolce
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | | | - Massimo Locati
- IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
7
|
Pepe G, Sfogliarini C, Rizzello L, Battaglia G, Pinna C, Rovati G, Ciana P, Brunialti E, Mornata F, Maggi A, Locati M, Vegeto E. ERα-independent NRF2-mediated immunoregulatory activity of tamoxifen. Biomed Pharmacother 2021; 144:112274. [PMID: 34653752 DOI: 10.1016/j.biopha.2021.112274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
Sex differences in immune-mediated diseases are linked to the activity of estrogens on innate immunity cells, including macrophages. Tamoxifen (TAM) is a selective estrogen receptor modulator (SERM) used in estrogen receptor-alpha (ERα)-dependent breast cancers and off-target indications such as infections, although the immune activity of TAM and its active metabolite, 4-OH tamoxifen (4HT), is poorly characterized. Here, we aimed at investigating the endocrine and immune activity of these SERMs in macrophages. Using primary cultures of female mouse macrophages, we analyzed the expression of immune mediators and activation of effector functions in competition experiments with SERMs and 17β-estradiol (E2) or the bacterial endotoxin LPS. We observed that 4HT and TAM induce estrogen antagonist effects when used at nanomolar concentrations, while pharmacological concentrations that are reached by TAM in clinical settings regulate the expression of VEGFα and other immune activation genes by ERα- and G protein-coupled receptor 1 (GPER1)-independent mechanisms that involve NRF2 through PI3K/Akt-dependent mechanisms. Importantly, we observed that SERMs potentiate cell phagocytosis and modify the effects of LPS on the expression of inflammatory cytokines, such as TNFα and IL1β, with an overall increase in cell inflammatory phenotype, further sustained by potentiation of IL1β secretion through caspase-1 activation. Altogether, our data unravel a novel molecular mechanism and immune functions for TAM and 4HT, sustaining their repurposing in infective and other estrogen receptors-unrelated pathologies.
Collapse
Affiliation(s)
- Giovanna Pepe
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Chiara Sfogliarini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; National Institute of Molecular Genetics (INGM) Milan, 20122 Milan, Italy
| | - Giuseppe Battaglia
- Department of Chemistry and; The EPSRC/Jeol Centre for Liquid Phase Electron Microscopy, University College London, WC1H 0AJ London, U.K; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Institute for Physics of Living System, University College London, WC1E 6BT London, U.K; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Christian Pinna
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Paolo Ciana
- Department of Health Sciences University of Milan, 20142 Milan, Italy
| | - Electra Brunialti
- Department of Health Sciences University of Milan, 20142 Milan, Italy
| | - Federica Mornata
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Adriana Maggi
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Massimo Locati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
8
|
Liao J, Yang F, Bai Y, Yu W, Qiao N, Han Q, Zhang H, Guo J, Hu L, Li Y, Pan J, Tang Z. Metabolomics analysis reveals the effects of copper on mitochondria-mediated apoptosis in kidney of broiler chicken (Gallus gallus). J Inorg Biochem 2021; 224:111581. [PMID: 34419760 DOI: 10.1016/j.jinorgbio.2021.111581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/30/2021] [Accepted: 08/11/2021] [Indexed: 01/01/2023]
Abstract
Copper (Cu) is one of the ubiquitous environmental pollutants which have raised wide concerns about the potential toxic effects and public health threat. For deeply investigating the nephrotoxicity induced by Cu, the effects of Cu on mitochondria-mediated apoptosis in kidney were first to analyze by combining metabolomics and molecular biology techniques. In this study, broiler chicks were fed with different contents of Cu (11, 110, 220, and 330 mg/kg Cu) for 49 d. The results of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining and transmission electron microscope showed that Cu could induce apoptosis in kidney, characterized by the increasing of TUNEL-positive cells and mitochondrial vacuolation. Additionally, a total of 62 differential metabolites were detected by liquid chromatography-mass spectrometry (LC-MS), and mainly enriched in the metabolic pathways including riboflavin metabolism, glutathione metabolism, sphingolipid metabolism, and glycerophospholipid metabolism, which were closely to mitochondrial metabolism. Meanwhile, the decreased mitochondrial membrane potential (MMP), increased mitochondrial membrane permeability and the change of mRNA and protein expression levels associated with mitochondria-mediated apoptosis and mitochondrial dynamics confirmed that Cu could induce mitochondria-mediated apoptosis. Therefore, our results demonstrated that Cu induced mitochondria-mediated apoptosis in kidney. Moreover, this study highlighted the metabolic characteristics of Cu to kidney, which suggested that mitochondrial metabolism could be considered as an important factor influencing toxicity.
Collapse
Affiliation(s)
- Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, PR China
| | - Yuman Bai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Wenlan Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Na Qiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
9
|
Liu J, Fan Y, Yu H, Xu T, Zhang C, Zhou L, Li G, Zhang Y. Allopurinol Protects Against Cholestatic Liver Injury in Mice Not Through Depletion of Uric Acid. Toxicol Sci 2021; 181:295-305. [PMID: 33749747 DOI: 10.1093/toxsci/kfab034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cholestasis is one of the most severe manifestations of liver injury and has limited therapeutic options. Allopurinol (AP), an inhibitor of uric acid (UA) synthesis, was reported to prevent liver damage in several liver diseases. However, whether AP protects against intrahepatic cholestatic liver injury and what is the role of UA in the pathogenesis of cholestasis remain unknown. In this study, we reported that AP attenuated liver injury in a mouse model of intrahepatic cholestasis induced by alpha-naphthylisothiocyanate (ANIT). AP showed no significant effect on glutathione depletion, inflammation, or bile acid metabolism in livers of ANIT-treated mice. Instead, AP significantly improved fatty acid β-oxidation in livers of ANIT-treated mice, which was associated with activation of PPARα. The protective effect of AP on cholestatic liver injury was not attributable to the depletion of UA, because both exogenous and endogenous UA prevented liver injury in ANIT-treated mice via inhibition of NF-kB-mediated inflammation. In conclusion, the present study provides a new perspective for the therapeutic use of AP and the role of UA in cholestatic liver injury.
Collapse
Affiliation(s)
- Jing Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Hang Yu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Tong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China
| | - Lijun Zhou
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Gentao Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China.,School of Pharmacy, Weifang Medical University, Shandong 261053, China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
10
|
Mitochondrial dysfunction and apoptosis underlie the hepatotoxicity of perhexiline. Toxicol In Vitro 2020; 69:104987. [PMID: 32861758 DOI: 10.1016/j.tiv.2020.104987] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/29/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
Perhexiline is an anti-anginal drug developed in the late 1960s. Despite its therapeutic success, it caused severe hepatoxicity in selective patients, which resulted in its withdrawal from the market. In the current study we explored the molecular mechanisms underlying the cytotoxicity of perhexiline. In primary human hepatocytes, HepaRG cells, and HepG2 cells, perhexiline induced cell death in a concentration- and time-dependent manner. Perhexiline treatment also caused a significant increase in caspase 3/7 activity at 2 h and 4 h. Pretreatment with specific caspase inhibitors suggested that both intrinsic and extrinsic apoptotic pathways contributed to perhexiline-induced cytotoxicity, which was confirmed by increased expression of TNF-α, cleavage of caspase 3 and 9 upon perhexiline treatment. Moreover, perhexiline caused mitochondrial dysfunction, demonstrated by the classic glucose-galactose assay at 4 h and 24 h. Results from JC-1 staining suggested perhexiline caused loss of mitochondrial potential. Blocking mitochondrial permeability transition pore using inhibitor bongkrekic acid attenuated the cytotoxicity of perhexiline. Western blotting analysis also showed decreased expression level of pro-survival proteins Bcl-2 and Mcl-1, and increased expression of pro-apoptotic protein Bad. Direct measurement of the activity of individual components of the mitochondrial respiratory complex demonstrated that perhexiline strongly inhibited Complex IV and Complex V and moderately inhibited Complex II and Complex II + III. Overall, our data demonstrated that both mitochondrial dysfunction and apoptosis underlies perhexiline-induced hepatotoxicity.
Collapse
|
11
|
Zainal-Abidin MH, Hayyan M, Ngoh GC, Wong WF, Looi CY. Potentiating the anti-cancer profile of tamoxifen-loaded graphene using deep eutectic solvents as functionalizing agents. APPLIED NANOSCIENCE 2019. [DOI: 10.1007/s13204-019-01105-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
12
|
Nerush AS, Shсhukina KM, Balalaeva IV, Orlova AG. Hydrogen peroxide in the reactions of cancer cells to cisplatin. Biochim Biophys Acta Gen Subj 2019; 1863:692-702. [PMID: 30690121 DOI: 10.1016/j.bbagen.2019.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/05/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hydrogen peroxide (H2O2) is thought to be one of the key components involved in the responses of tumor cells to chemotherapy. The aim of this study was to reveal the pathways and the phases of cisplatin-induced cell death that are characterized by changes of H2O2 level. METHODS The genetically encoded cytosolic fluorescent sensor HyPer2 was used for flow cytometric analysis of the cisplatin-induced changes in H2O2 level in HeLa Kyoto cells. Using a vital dye and the apoptotic markers PE Annexin V or TMRE the pathways and stages of cell death were investigated simultaneously with HyPer2 response. The H2O2 level was studied separately in viable and early apoptotic cells after 12, 18, 24 h's incubation with cisplatin at several concentrations with or without the scavenger of reactive oxygen species NAC. RESULTS Cisplatin causes dose- and time-dependent increase of H2O2 level in TMRE-positive and PE Annexin V-negative cancer cells. The scavenging of ROS by NAC decreased the H2O2 level and restored cell viability. CONCLUSION Н2О2 generation begins in cells that have already lost mitochondrial membrane potential but have not yet externalized phosphatidylserine. Prevention of apoptosis by NAC confirmed the role of H2O2 in apoptosis induction. GENERAL SIGNIFICANCE This is the first time that the sensor HyPer2 has been used in parallel with apoptotic markers and vital dye to demonstrate the role of H2O2 in different stages and types of tumor cell death under chemotherapeutic action.
Collapse
Affiliation(s)
- A S Nerush
- Institute of Applied Physics RAS, Nizhny Novgorod, Russia.
| | - K M Shсhukina
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - I V Balalaeva
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - A G Orlova
- Institute of Applied Physics RAS, Nizhny Novgorod, Russia
| |
Collapse
|
13
|
Poly-ADP-Ribosylation of Estrogen Receptor-Alpha by PARP1 Mediates Antiestrogen Resistance in Human Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11010043. [PMID: 30621214 PMCID: PMC6357000 DOI: 10.3390/cancers11010043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Therapeutic targeting of estrogen receptor-α (ERα) by the anti-estrogen tamoxifen is standard of care for premenopausal breast cancer patients and remains a key component of treatment strategies for postmenopausal patients. While tamoxifen significantly increases overall survival, tamoxifen resistance remains a major limitation despite continued expression of ERα in resistant tumors. Previous reports have described increased oxidative stress in tamoxifen resistant versus sensitive breast cancer and a role for PARP1 in mediating oxidative damage repair. We hypothesized that PARP1 activity mediated tamoxifen resistance in ERα-positive breast cancer and that combining the antiestrogen tamoxifen with a PARP1 inhibitor (PARPi) would sensitize tamoxifen resistant cells to tamoxifen therapy. In tamoxifen-resistant vs. -sensitive breast cancer cells, oxidative stress and PARP1 overexpression were increased. Furthermore, differential PARylation of ERα was observed in tamoxifen-resistant versus -sensitive cells, and ERα PARylation was increased by tamoxifen treatment. Loss of ERα PARylation following treatment with a PARP inhibitor (talazoparib) augmented tamoxifen sensitivity and decreased localization of both ERα and PARP1 to ERα-target genes. Co-administration of talazoparib plus tamoxifen increased DNA damage accumulation and decreased cell survival in a dose-dependent manner. The ability of PARPi to overcome tamoxifen resistance was dependent on ERα, as lack of ERα-mediated estrogen signaling expression and showed no response to tamoxifen-PARPi treatment. These results correlate ERα PARylation with tamoxifen resistance and indicate a novel mechanism-based approach to overcome tamoxifen resistance in ER+ breast cancer.
Collapse
|
14
|
Sun MY, Song YN, Zhang M, Zhang CY, Zhang LJ, Zhang H. Ginsenoside Rg3 inhibits the migration and invasion of liver cancer cells by increasing the protein expression of ARHGAP9. Oncol Lett 2018; 17:965-973. [PMID: 30655855 PMCID: PMC6313058 DOI: 10.3892/ol.2018.9701] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/10/2018] [Indexed: 12/13/2022] Open
Abstract
Ginsenoside Rg3, a naturally occurring phytochemical, serves an important role in the prevention and treatment of cancer. In the present study, with the aim to reveal the molecular mechanism of Rg3 in liver cancer cell metastasis, the anti-migration and anti-invasion effects of Rg3 on liver cancer cells were investigated. It was demonstrated that Rg3 caused marked inhibition of cell migration and invasion of human liver cancer cells, HepG2 and MHCC-97L, in vitro, and the growth of HepG2 and MHCC-97L tumors in BABL/c nude mice. The protein expression of Rho GTPase activating protein 9 (ARHGAP9) was increased both in HepG2 and MHCC-97L cells. Following ARHGAP9 knockdown, the results of Transwell and tumorigenesis assays revealed that the anti-migration, anti-invasion and anti-tumor growth effects of Rg3 were impaired significantly. The increased expression of ARHGAP9 protein induced by Rg3 was remarkably suppressed. All results suggested that ARHGAP9 protein may be a vital regulator in the anti-metastatic role of Rg3. To the best of our knowledge, the present study is the first to report that Rg3 effectively suppressed the migration and invasion of liver cancer cells by upregulating the protein expression of ARHGAP9, indicating a novel natural therapeutic agent and a therapeutic target for the treatment of liver cancer.
Collapse
Affiliation(s)
- Meng-Yao Sun
- Department of Pharmaceutical Botany, School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Ya-Nan Song
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Miao Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Chun-Yan Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Li-Jun Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Hong Zhang
- Department of Pharmaceutical Botany, School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China.,Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| |
Collapse
|
15
|
Joshi P, Kang SY, Datar A, Lee MY. High-Throughput Assessment of Mechanistic Toxicity of Chemicals in Miniaturized 3D Cell Culture. ACTA ACUST UNITED AC 2018; 79:e66. [PMID: 30387930 DOI: 10.1002/cptx.66] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High-content imaging (HCI) assays on two-dimensional (2D) cell cultures often do not represent in vivo characteristics accurately, thus reducing the predictability of drug toxicity/efficacy in vivo. On the other hand, conventional 3D cell cultures are relatively low throughput and possess difficulty in cell imaging. To address these limitations, a miniaturized 3D cell culture has been developed on a micropillar/microwell chip platform with human cells encapsulated in biomimetic hydrogels. Model compounds are used to validate human cell microarrays for high-throughput assessment of mechanistic toxicity. Main mechanisms of toxicity of compounds can be investigated by analyzing multiple parameters such as DNA damage, mitochondrial impairment, intracellular glutathione level, and cell membrane integrity. IC50 values of these parameters can be determined and compared for the compounds to investigate the main mechanism of toxicity. This paper describes miniaturized HCI assays on 3D-cultured cell microarrays for high-throughput assessment of mechanistic profiles of compound-induced toxicity. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Pranav Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Soo-Yeon Kang
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Akshata Datar
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Moo-Yeal Lee
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
16
|
Joshi P, Datar A, Yu KN, Kang SY, Lee MY. High-content imaging assays on a miniaturized 3D cell culture platform. Toxicol In Vitro 2018; 50:147-159. [PMID: 29501531 DOI: 10.1016/j.tiv.2018.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
The majority of high-content imaging (HCI) assays have been performed on two-dimensional (2D) cell monolayers for its convenience and throughput. However, 2D-cultured cell models often do not represent the in vivo characteristics accurately and therefore reduce the predictability of drug toxicity/efficacy in vivo. Recently, three-dimensional (3D) cell-based HCI assays have been demonstrated to improve predictability, but its use is limited due to difficulty in maneuverability and low throughput in cell imaging. To alleviate these issues, we have developed miniaturized 3D cell culture on a micropillar/microwell chip and demonstrated high-throughput HCI assays for mechanistic toxicity. Briefly, Hep3B human hepatoma cell line was encapsulated in a mixture of alginate and fibrin gel on the micropillar chip, cultured in 3D, and exposed to six model compounds in the microwell chip for rapidly assessing mechanistic hepatotoxicity. Several toxicity parameters, including DNA damage, mitochondrial impairment, intracellular glutathione level, and cell membrane integrity were measured on the chip, and the IC50 values of the compounds at different readouts were determined to investigate the mechanism of toxicity. Overall, the Z' factors were between 0.6 and 0.8 for the HCI assays, and the coefficient of variation (CV) were below 20%. These results indicate high robustness and reproducibility of the HCI assays established on the miniaturized 3D cell culture chip. In addition, it was possible to determine the predominant mechanism of toxicity using the 3D HCI assays. Therefore, our miniaturized 3D cell culture coupled with HCI assays has great potential for high-throughput screening (HTS) of compounds and mechanistic toxicity profiling.
Collapse
Affiliation(s)
- Pranav Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Akshata Datar
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Kyeong-Nam Yu
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Soo-Yeon Kang
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Moo-Yeal Lee
- Department of Chemical and Biomedical Engineering, Cleveland State University, 455 Fenn Hall, 1960 East 24th Street, Cleveland, OH 44115-2214, USA.
| |
Collapse
|
17
|
Sanaei M, Kavoosi F, Atashpour S, Haghighat S. Effects of Genistein and Synergistic Action in Combination with Tamoxifen on the HepG2 Human Hepatocellular Carcinoma Cell Line. Asian Pac J Cancer Prev 2017; 18:2381-2385. [PMID: 28950682 PMCID: PMC5720640 DOI: 10.22034/apjcp.2017.18.9.2381] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Introduction: The flavonoids comprise a diverse group of polyphenolic compounds with antioxidant activity that is present in edible plants like soybeans and soy products. In vivo studies have concentrated on the effects of flavonoids on cancer and genistein (GE), a soy-derived isoflavone, has been reported to reduce prostate, colon, hepatic and breast adenocarcinoma risk. Tamoxifen (TAM) is an important drug for cancer treatment worldwide, which can induce apoptosis in various cancers, including examples in the liver, breast and ovaries. The aim of the present study was to evaluate the effects of GE and TAM, alone and in combination, on proliferation and apoptosis in the human hepatocellular carcinoma (HCC) HepG2 cell line. Materials and Methods: HepG 2 cells were treated with GE, TAM and GE/TAM and then MTT and flow cytometry assays were conducted to determine effects on viability and apoptosis, respectively. Results: GE and TAM inhibited cell proliferation and induced apoptosis in the HepG 2 cell lines. Discussion: Our findings clearly indicated that GE and TAM may exert inhibitory and apoptotic effects in liver cancer cells. Conclusion: GE and TAM can significantly inhibit growth of HCC cells and play a significant role in apoptosis.
Collapse
Affiliation(s)
- Masumeh Sanaei
- Research Center for Non-communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | | | | | | |
Collapse
|
18
|
Chiaravalli J, Glickman JF. A High-Content Live-Cell Viability Assay and Its Validation on a Diverse 12K Compound Screen. SLAS DISCOVERY 2017; 22:1120-1130. [DOI: 10.1177/2472555217724745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have developed a new high-content cytotoxicity assay using live cells, called “ImageTOX.” We used a high-throughput fluorescence microscope system, image segmentation software, and the combination of Hoechst 33342 and SYTO 17 to simultaneously score the relative size and the intensity of the nuclei, the nuclear membrane permeability, and the cell number in a 384-well microplate format. We then performed a screen of 12,668 diverse compounds and compared the results to a standard cytotoxicity assay. The ImageTOX assay identified similar sets of compounds to the standard cytotoxicity assay, while identifying more compounds having adverse effects on cell structure, earlier in treatment time. The ImageTOX assay uses inexpensive commercially available reagents and facilitates the use of live cells in toxicity screens. Furthermore, we show that we can measure the kinetic profile of compound toxicity in a high-content, high-throughput format, following the same set of cells over an extended period of time.
Collapse
Affiliation(s)
- Jeanne Chiaravalli
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - J. Fraser Glickman
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY, USA
| |
Collapse
|
19
|
Zhang R, Ai X, Duan Y, Xue M, He W, Wang C, Xu T, Xu M, Liu B, Li C, Wang Z, Zhang R, Wang G, Tian S, Liu H. Kaempferol ameliorates H9N2 swine influenza virus-induced acute lung injury by inactivation of TLR4/MyD88-mediated NF-κB and MAPK signaling pathways. Biomed Pharmacother 2017; 89:660-672. [DOI: 10.1016/j.biopha.2017.02.081] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 01/23/2023] Open
|
20
|
Radin DP, Patel P. Delineating the molecular mechanisms of tamoxifen’s oncolytic actions in estrogen receptor-negative cancers. Eur J Pharmacol 2016; 781:173-80. [DOI: 10.1016/j.ejphar.2016.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/06/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022]
|
21
|
Bekele RT, Venkatraman G, Liu RZ, Tang X, Mi S, Benesch MGK, Mackey JR, Godbout R, Curtis JM, McMullen TPW, Brindley DN. Oxidative stress contributes to the tamoxifen-induced killing of breast cancer cells: implications for tamoxifen therapy and resistance. Sci Rep 2016; 6:21164. [PMID: 26883574 PMCID: PMC4756695 DOI: 10.1038/srep21164] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023] Open
Abstract
Tamoxifen is the accepted therapy for patients with estrogen receptor-α (ERα)-positive breast cancer. However, clinical resistance to tamoxifen, as demonstrated by recurrence or progression on therapy, is frequent and precedes death from metastases. To improve breast cancer treatment it is vital to understand the mechanisms that result in tamoxifen resistance. This study shows that concentrations of tamoxifen and its metabolites, which accumulate in tumors of patients, killed both ERα-positive and ERα-negative breast cancer cells. This depended on oxidative damage and anti-oxidants rescued the cancer cells from tamoxifen-induced apoptosis. Breast cancer cells responded to tamoxifen-induced oxidation by increasing Nrf2 expression and subsequent activation of the anti-oxidant response element (ARE). This increased the transcription of anti-oxidant genes and multidrug resistance transporters. As a result, breast cancer cells are able to destroy or export toxic oxidation products leading to increased survival from tamoxifen-induced oxidative damage. These responses in cancer cells also occur in breast tumors of tamoxifen-treated mice. Additionally, high levels of expression of Nrf2, ABCC1, ABCC3 plus NAD(P)H dehydrogenase quinone-1 in breast tumors of patients at the time of diagnosis were prognostic of poor survival after tamoxifen therapy. Therefore, overcoming tamoxifen-induced activation of the ARE could increase the efficacy of tamoxifen in treating breast cancer.
Collapse
Affiliation(s)
- Raie T Bekele
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Ganesh Venkatraman
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Rong-Zong Liu
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Xiaoyun Tang
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Si Mi
- Department of Agricultural, Food and Nutritional Science (Lipid Chemistry Group), University of Alberta, Edmonton, Alberta, T6G 2P5, Canada
| | - Matthew G K Benesch
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - John R Mackey
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Roseline Godbout
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science (Lipid Chemistry Group), University of Alberta, Edmonton, Alberta, T6G 2P5, Canada
| | - Todd P W McMullen
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada.,Department of Surgery, Walter C Mackenzie Health Science Centre, University of Alberta, Edmonton, T6G 2R7, Alberta, Canada
| | - David N Brindley
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| |
Collapse
|
22
|
Guo CH, Hsia S, Hsiung DY, Chen PC. Supplementation with Selenium yeast on the prooxidant-antioxidant activities and anti-tumor effects in breast tumor xenograft-bearing mice. J Nutr Biochem 2015; 26:1568-79. [PMID: 26344777 DOI: 10.1016/j.jnutbio.2015.07.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/26/2015] [Accepted: 07/29/2015] [Indexed: 12/29/2022]
Abstract
Selenium (Se) is essential for antioxidant activity involved in immune function and anti-carcinogenic action, whereas at higher concentrations, Se may have pro-oxidant properties. The present study was aimed at determining the effects of Se supplementation, as Se yeast, on oxidative stress in non-tumor/tumor tissues, as well as regulation of the apoptotic process, and immune responses in mice-bearing breast tumor xenografts. Female BALB/cByJNarl mice were divided into control (CNL and CNL-con), Se-supplemented control (CNL-HS, given as a single oral dose of 912 ng Se daily), breast tumor-bearing (TB and TB-con), TB-LS (228 ng Se), TB-MS (456 ng Se) and TB-HS (912 ng Se) groups. All mice were treated with/without Se for 14 days. A number of variables were further measured. Compared with the TB groups, tumor bearing mice with Se supplement had increased plasma Se concentrations, reduced erythrocyte Se-dependent glutathione peroxidase (GPx) activity and malondialdehyde (MDA) products and inhibited tumor growth. They have also higher Se concentrations in non-tumor and tumor tissues. Significantly elevated concentrations of MDA and reduced GPx activities, as well as increased anti-apoptotic bcl-2 and tumor suppressor p53 concentrations in tumor tissues were observed as Se accumulated in tumor, whereas lower MDA products were found in various non-tumor tissues than did the corresponding values. Further, there were elevated concentrations of Th1-derived cytokines and decreased Th2-type interleukin (IL)-4 in tumor-bearing mice with the treatment of Se. In conclusion, accumulation of Se in tumors may induce oxidative stress and p53-dependent pro-oxidative apoptosis, thus inhibiting the growth of breast tumor.
Collapse
Affiliation(s)
- Chih-Hung Guo
- Institute of Biomedical Nutrition, Hung-Kuang University, Taichung, 433, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, 413, Taiwan; Taiwan Nutraceutical Association, Taipei 115, Taiwan.
| | - Simon Hsia
- Institute of Biomedical Nutrition, Hung-Kuang University, Taichung, 433, Taiwan; Taiwan Nutraceutical Association, Taipei 115, Taiwan
| | - Der-Yun Hsiung
- Department of Nursing, Hung-Kuang University, Taichung, 404, Taiwan; School of Nursing, China Medical University, Taichung, 404, Taiwan
| | - Pei-Chung Chen
- Taiwan Nutraceutical Association, Taipei 115, Taiwan; College of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan; College of Engineering, National Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
23
|
Xu T, Wang C, Zhang R, Xu M, Liu B, Wei D, Wang G, Tian S. Carnosine markedly ameliorates H9N2 swine influenza virus-induced acute lung injury. J Gen Virol 2015; 96:2939-2950. [PMID: 26233716 DOI: 10.1099/jgv.0.000238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Oxidative stress injury is an important pathogenesis of influenza virus in critically ill patients. The present study investigated the efficacy of carnosine, an antioxidant and free radical scavenger, on a model of acute lung injury (ALI) induced by H9N2 swine influenza virus. Female specific-pathogen-free BALB/c mice were randomized into four groups and treated as follows: (1) H9N2 group, (2) mock control group, (3) H9N2+carnosine group and (4) carnosine control group. The H9N2 group mice were inoculated intranasally with A/Swine/Hebei/012/2008/ (H9N2) virus (100 μl) in allantoic fluid (AF), whilst mock-infected animals were intranasally inoculated with non-infectious AF. Carnosine [10 mg (kg body mass)- 1] was administered orally (100 μl) for 7 days consecutively. The survival rate, lung water content, TNF-α and IL-1β levels, lung histopathology, myeloperoxidase (MPO) activity, and Toll-like receptor (TLR)-4 levels were determined at 2, 4, 6, 8 and 14 days after inoculation. Carnosine treatment effectively decreased the mortality (43 versus 75 %, P < 0.05), significantly ameliorated pathological lesions in lungs and decreased the lung wet/dry mass ratio (P < 0.05). It also inhibited MPO activity, suppressed TNF-α and IL-1β release, decreased the H9N2 viral titre, and markedly inhibited levels of TLR-4 mRNA and protein in the lungs of infected mice (P < 0.05), which supported the use of carnosine for managing severe influenza cases.
Collapse
Affiliation(s)
- Tong Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Cunlian Wang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Ruihua Zhang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Mingju Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Baojian Liu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Dong Wei
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Guohua Wang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| | - Shufei Tian
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, Hebei, PR China
| |
Collapse
|
24
|
Jena SK, Suresh S, Sangamwar AT. Modulation of tamoxifen-induced hepatotoxicity by tamoxifen-phospholipid complex. ACTA ACUST UNITED AC 2015; 67:1198-206. [PMID: 25904227 DOI: 10.1111/jphp.12422] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/08/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Tamoxifen (TMX), a non-steroidal antiestrogen is a first-line drug in the treatment and prevention of all stages of estrogen-receptor-positive breast cancer. However, oxidative liver damage and hepatocarcinoma are the major problems associated with its long-term clinical use. The aim of this study was to investigate the ameliorative effect of phospholipid against TMX-induced hepatotoxicity. METHODS Fifteen female Sprague-Dawley rats were divided into three groups with five rats in each group. Group I received only standard diet and distilled water for 28 days and served as normal. Group II received TMX per day p.o., for 28 days and served as control, and group III received TMX-phospholipid complex (TMX-PLC) per day p.o., for 28 days. Rats were examined for the effect of phospholipid on TMX-induced depletion of antioxidant enzymes, serum biochemical parameters and induction of lipid peroxidation. KEY FINDINGS Treatment with TMX-PLC significantly ameliorates the TMX-induced hepatotoxicity by diminishing the toxicity markers such lipid peroxidation, aspartate transaminase and alanine transaminase, accompanied by an increase in antioxidant enzyme activity in TMX-treated rats. Histological findings further confirmed the hepatoprotective effect of phospholipid. CONCLUSIONS Data of the present study suggests that phospholipid may prove as a useful component of combination therapy in cancer patients under the TMX treatment regimen.
Collapse
Affiliation(s)
- Sunil K Jena
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, India
| | - Sarasija Suresh
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, India
| |
Collapse
|
25
|
Leinonen HM, Kansanen E, Pölönen P, Heinäniemi M, Levonen AL. Role of the Keap1-Nrf2 pathway in cancer. Adv Cancer Res 2015; 122:281-320. [PMID: 24974185 DOI: 10.1016/b978-0-12-420117-0.00008-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor E2-related factor 2 (Nrf2) pathway is one of the major signaling cascades involved in cell defense and survival against endogenous and exogenous stress. While Nrf2 and its target genes provide protection against various age-related diseases including tumorigenesis, constitutively active Nrf2 in cancer cells increases the expression of cytoprotective genes and, consequently, enhances proliferation via metabolic reprogramming and inhibition of apoptosis. Herein, we review the current understanding of the regulation of Nrf2 in normal cells as well as its dual role in cancer. Furthermore, the mechanisms of Nrf2 dysregulation in cancer, consequences of unchecked Nrf2 activity, and therapies targeting the Keap1-Nrf2 system are discussed.
Collapse
Affiliation(s)
- Hanna M Leinonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland
| | - Emilia Kansanen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland
| | - Petri Pölönen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland; Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland
| | - Merja Heinäniemi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland; Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland
| | - Anna-Liisa Levonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland.
| |
Collapse
|
26
|
Tamoxifen reduces fat mass by boosting reactive oxygen species. Cell Death Dis 2015; 6:e1586. [PMID: 25569103 PMCID: PMC4669751 DOI: 10.1038/cddis.2014.553] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/10/2014] [Accepted: 11/17/2014] [Indexed: 01/04/2023]
Abstract
As the pandemic of obesity is growing, a variety of animal models have been generated to study the mechanisms underlying the increased adiposity and development of metabolic disorders. Tamoxifen (Tam) is widely used to activate Cre recombinase that spatiotemporally controls target gene expression and regulates adiposity in laboratory animals. However, a critical question remains as to whether Tam itself affects adiposity and possibly confounds the functional study of target genes in adipose tissue. Here we administered Tam to Cre-absent forkhead box O1 (FoxO1) floxed mice (f-FoxO1) and insulin receptor substrate Irs1/Irs2 double floxed mice (df-Irs) and found that Tam induced approximately 30% reduction (P<0.05) in fat mass with insignificant change in body weight. Mechanistically, Tam promoted reactive oxygen species (ROS) production, apoptosis and autophagy, which was associated with downregulation of adipogenic regulator peroxisome proliferator-activated receptor gamma and dedifferentiation of mature adipocytes. However, normalization of ROS potently suppressed Tam-induced apoptosis, autophagy and adipocyte dedifferentiation, suggesting that ROS may account, at least in part, for the changes. Importantly, Tam-induced ROS production and fat mass reduction lasted for 4–5 weeks in the f-FoxO1 and df-Irs mice. Our data suggest that Tam reduces fat mass via boosting ROS, thus making a recovery period crucial for posttreatment study.
Collapse
|
27
|
Jennings P, Schwarz M, Landesmann B, Maggioni S, Goumenou M, Bower D, Leonard MO, Wiseman JS. SEURAT-1 liver gold reference compounds: a mechanism-based review. Arch Toxicol 2014; 88:2099-133. [DOI: 10.1007/s00204-014-1410-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/01/2014] [Indexed: 12/20/2022]
|
28
|
Yaacob NS, Ismail NF. Comparison of cytotoxicity and genotoxicity of 4-hydroxytamoxifen in combination with Tualang honey in MCF-7 and MCF-10A cells. Altern Ther Health Med 2014; 14:106. [PMID: 24646375 PMCID: PMC3994783 DOI: 10.1186/1472-6882-14-106] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 03/12/2014] [Indexed: 02/07/2023]
Abstract
Background The Malaysian Tualang honey (TH) is not only cytotoxic to human breast cancer cell lines but it has recently been reported to promote the anticancer activity induced by tamoxifen in MCF-7 and MDA-MB-231 cells suggesting its potential as an adjuvant for the chemotherapeutic agent. However, tamoxifen produces adverse effects that could be due to its ability to induce cellular DNA damage. Therefore, the study is undertaken to determine the possible modulation of the activity of 4-hydroxytamoxifen (OHT), an active metabolite of tamoxifen, by TH in non-cancerous epithelial cell line, MCF-10A, in comparison with MCF-7 cells. Methods MCF-7 and MCF-10A cells were treated with TH, OHT or the combination of both and cytotoxicity and antiproliferative activity were determined using LDH and MTT assays, respectively. The effect on cellular DNA integrity was analysed by comet assay and the expression of DNA repair enzymes was determined by Western blotting. Results OHT exposure was cytotoxic to both cell lines whereas TH was cytotoxic to MCF-7 cells only. TH also significantly decreased the cytotoxic effect of OHT in MCF-10A but not in MCF-7 cells. TH induced proliferation of MCF10A cells but OHT caused growth inhibition that was abrogated by the concomitant treatment with TH. While TH enhanced the OHT-induced DNA damage in the cancer cells, it dampened the genotoxic effect of OHT in the non-cancerous cells. This was supported by the increased expression of DNA repair proteins, Ku70 and Ku80, in MCF-10A cells by TH. Conclusion The findings indicate that TH could afford protection of non-cancerous cells from the toxic effects of tamoxifen by increasing the efficiency of DNA repair mechanism in these cells.
Collapse
|
29
|
Segala G, de Medina P, Iuliano L, Zerbinati C, Paillasse MR, Noguer E, Dalenc F, Payré B, Jordan VC, Record M, Silvente-Poirot S, Poirot M. 5,6-Epoxy-cholesterols contribute to the anticancer pharmacology of tamoxifen in breast cancer cells. Biochem Pharmacol 2013; 86:175-89. [PMID: 23500540 DOI: 10.1016/j.bcp.2013.02.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/21/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Tamoxifen (Tam) is a selective estrogen receptor modulator (SERM) that remains one of the major drugs used in the hormonotherapy of breast cancer (BC). In addition to its SERM activity, we recently showed that the oxidative metabolism of cholesterol plays a role in its anticancer pharmacology. We established that these effects were not regulated by the ER but by the microsomal antiestrogen binding site/cholesterol-5,6-epoxide hydrolase complex (AEBS/ChEH). The present study aimed to identify the oxysterols that are produced under Tam treatment and to define their mechanisms of action. Tam and PBPE (a selective AEBS/ChEH ligand) stimulated the production and the accumulation of 5,6α-epoxy-cholesterol (5,6α-EC), 5,6α-epoxy-cholesterol-3β-sulfate (5,6-ECS), 5,6β-epoxy-cholesterol (5,6β-EC) in MCF-7 cells through a ROS-dependent mechanism, by inhibiting ChEH and inducing sulfation of 5,6α-EC by SULT2B1b. We showed that only 5,6α-EC was responsible for the induction of triacylglycerol (TAG) biosynthesis by Tam and PBPE, through the modulation of the oxysterol receptor LXRβ. The cytotoxicity mediated by Tam and PBPE was triggered by 5,6β-EC through an LXRβ-independent route and by 5,6-ECS through an LXRβ-dependent mechanism. The importance of SULT2B1b was confirmed by its ectopic expression in the SULT2B1b(-) MDA-MB-231 cells, which became sensitive to 5,6α-EC, Tam or PBPE at a comparable level to MCF-7 cells. This study established that 5,6-EC metabolites contribute to the anticancer pharmacology of Tam and highlights a novel signaling pathway that points to a rationale for re-sensitizing BC cells to Tam and AEBS/ChEH ligands.
Collapse
Affiliation(s)
- Gregory Segala
- UMR 1037 INSERM-University Toulouse III, Cancer Research Center of Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Penney RB, Roy D. Thioredoxin-mediated redox regulation of resistance to endocrine therapy in breast cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:60-79. [PMID: 23466753 DOI: 10.1016/j.bbcan.2013.02.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 12/27/2022]
Abstract
Resistance to endocrine therapy in breast carcinogenesis due to the redox regulation of the signal transduction system by reactive oxygen species (ROS) is the subject of this review article. Both antiestrogens and aromatase inhibitors are thought to prevent cancer through modulating the estrogen receptor function, but other mechanisms cannot be ruled out as these compounds also block metabolism and redox cycling of estrogen and are free radical scavengers. Endocrine therapeutic agents, such as, tamoxifen and other antiestrogens, and the aromatase inhibitor, exemestane, are capable of producing ROS. Aggressive breast cancer cells have high oxidative stress and chronic treatment with exemestane, fulvestrant or tamoxifen may add additional ROS stress. Breast cancer cells receiving long-term antiestrogen treatment appear to adapt to this increased persistent level of ROS. This, in turn, may lead to the disruption of reversible redox signaling that involves redox-sensitive phosphatases, protein kinases, such as, ERK and AKT, and transcription factors, such as, AP-1, NRF-1 and NF-κB. Thioredoxin modulates the expression of estrogen responsive genes through modulating the production of H2O2 in breast cancer cells. Overexpressing thioredoxine reductase 2 and reducing oxidized thioredoxin restores tamoxifen sensitivity to previously resistant breast cancer cells. In summary, it appears that resistance to endocrine therapy may be mediated, in part, by ROS-mediated dysregulation of both estrogen-dependent and estrogen-independent redox-sensitive signaling pathways. Further studies are needed to define the mechanism of action of thioredoxin modifiers, and their effect on the redox regulation that contributes to restoring the antiestrogen-mediated signal transduction system and growth inhibitory action.
Collapse
Affiliation(s)
- Rosalind Brigham Penney
- Department of Environmental and Occupational Health, Florida International University, Miami, FL 33199, USA
| | | |
Collapse
|
31
|
Chakraborty A, Bodipati N, Demonacos MK, Peddinti R, Ghosh K, Roy P. Long term induction by pterostilbene results in autophagy and cellular differentiation in MCF-7 cells via ROS dependent pathway. Mol Cell Endocrinol 2012; 355:25-40. [PMID: 22273805 DOI: 10.1016/j.mce.2012.01.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 12/20/2011] [Accepted: 01/10/2012] [Indexed: 01/28/2023]
Abstract
This study shows the effect of pterostilbene on intracellular neutral lipid accumulation in MCF-7 breast cancer cells leading to growth arrest and autophagy. On exposing the breast cancer cells with 30 μM pterostilbene for 72 h there was almost 2-folds increase in neutral lipids and triglycerides. Also the phytochemical caused a 4-folds increase in the expression of adipogenic differentiation marker c/EBPα. Further, pterostilbene inhibited 3β-hydroxylsterol-Δ(7)-reductase, the enzyme which catalyzes the last step conversion of 7-dehydrocholesterol to cholesterol, and thereby causes the intracellular accumulation of the former sterol. These results were associated with over-expression of oxysterol binding protein homologue and liver X receptor (LXR) by ~7-folds. Pterostilbene also caused a simultaneous increase in the expression autophagic marker proteins Beclin 1 and LC3 II (microtubule-associated protein 1 light chain 3) by approximately 6-folds, which leads to an alternative pathway of autophagy. These effects were observed in association with the loss of mitotic and metastatic potential of MCF-7 cells which was abolished in the presence of catalase (ROS scavenger) or 3MA (autophagic inhibitor). Thus the present data shows that the long term exposure to pterostilbene causes growth arrest in MCF-7 cells which may be due to differentiation of the mammary carcinoma cells into normal epithelial cell like morphology and activation of autophagy.
Collapse
Affiliation(s)
- Ajanta Chakraborty
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | | | | | | | | | | |
Collapse
|
32
|
Tolosa L, Pinto S, Donato MT, Lahoz A, Castell JV, O’Connor JE, Gómez-Lechón MJ. Development of a Multiparametric Cell-based Protocol to Screen and Classify the Hepatotoxicity Potential of Drugs. Toxicol Sci 2012; 127:187-98. [DOI: 10.1093/toxsci/kfs083] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
33
|
Terashima J, Habano W, Gamou T, Ozawa S. Induction of CYP1 family members under low-glucose conditions requires AhR expression and occurs through the nuclear translocation of AhR. Drug Metab Pharmacokinet 2011; 26:577-83. [PMID: 21878739 DOI: 10.2133/dmpk.dmpk-11-rg-054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cross-talk between the aryl hydrocarbon receptor (AhR) pathway and the typical stress response is thought to be an important signal transduction in response to nutrient-stress conditions, such as glucose deprivation in liver cells. In the present study, we demonstrate that reduction of glucose concentration in the medium of HepG2 cells, a human hepatocellular carcinoma cell line, induces the CYP1 family and Nrf2. RNAi for AhR abolishes the induction of expression of CYP1 and Nrf2. These inductions are accompanied by the translocation of AhR into the nucleus in response to low-glucose conditions. Endogenous compounds are recruited as AhR ligands to induce various gene expressions, and our present results suggest that an endogenous AhR ligand is produced under low-glucose conditions and that the role of AhR as a transcription factor is related to the low-glucose response. The recommended glucose concentration (4.5 g/L) in the medium for culture of HepG2 was used as the high-glucose concentration in this study. We adopted 1.0 g/L as the low-glucose condition for elucidation of mechanisms of the stress response. These results will be useful to understand the relationship between drug-metabolizing enzymes and mechanisms of the anti-stress response of tumor cells, and will also be useful for investigating preventive remedies against tumor angiogenesis.
Collapse
Affiliation(s)
- Jun Terashima
- Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Japan.
| | | | | | | |
Collapse
|
34
|
Tamoxifen promotes superoxide production in platelets by activation of PI3-kinase and NADPH oxidase pathways. Thromb Res 2011; 129:36-42. [PMID: 21875743 DOI: 10.1016/j.thromres.2011.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 07/06/2011] [Accepted: 08/03/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tamoxifen is a selective estrogen receptor antagonist that is widely used for treatment and prevention of breast cancer. However, tamoxifen use can lead to an increased incidence of thrombotic events. The reason for this adverse event remains unknown. Previous studies showed that tamoxifen and its active metabolite Z-4-hydroxytamoxifen rapidly increased intracellular free calcium ([Ca(2+)](i)) in human platelets by a non-genomic mechanism that involved the activation of phospholipase C. Platelets play a pivotal role in thrombosis and Ca(2+) elevation is a central event in platelet activation. Therefore the mechanism by which tamoxifen activated Ca(2+) entry into platelets was investigated. METHODS [Ca(2+)](i) was measured using the fluorescent indicator fura-2 and reactive oxygen species were measured using lucigenin in isolated human platelets. RESULTS Tamoxifen analogs E-4-hydroxytamoxifen, with weak activity at the nuclear estrogen receptor and Z-4-hydroxytamoxifen, with strong activity at nuclear estrogen receptor, were equally active at increasing [Ca(2+)](i) and synergizing with ADP and thrombin to increase [Ca(2+)](i) in platelets. This result suggests that the effects of tamoxifen and E- and Z-4-hydroxytamoxifen to increase [Ca(2+)](i) are not mediated by the classical genomic estrogen receptor. The effects of tamoxifen to increase [Ca(2+)](i) were strongly inhibited by apocynin and apocynin dimer. This suggests that tamoxifen activates NADPH oxidase which leads to superoxide generation and in turn caused an increase in [Ca(2+)](i). Free radical scavengers TEMPO and TEMPOL also inhibited tamoxifen-induced [Ca(2+)](i) elevation. Inhibition of phosphoinositide-3-kinase (PI3-kinase), an upstream effector of NADPH oxidase with wortmannin and LY-294,002 also caused substantial inhibition of tamoxifen-induced elevation of [Ca(2+)](i). CONCLUSION Tamoxifen increases [Ca(2+)](i) in human platelets by a non-genomic mechanism. Tamoxifen activates phospholipase Cγ as well as PI3-kinase and NADPH oxidase pathway to generate superoxide which causes the release of Ca(2+) from the endoplasmic reticulum, and promotes Ca(2+) influx into the platelets.
Collapse
|
35
|
Yi L, Ji XX, Tan H, Lin M, Tang Y, Wen L, Ma YH, Su Q. Role of Ras-related C3 botulinum toxin substrate 2 (Rac2), NADPH oxidase and reactive oxygen species in diallyl disulphide-induced apoptosis of human leukaemia HL-60 cells. Clin Exp Pharmacol Physiol 2010; 37:1147-53. [PMID: 20804509 DOI: 10.1111/j.1440-1681.2010.05444.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
1. Diallyl disulphide (DADS) has potential as a chemopreventive and therapeutic agent. Previous studies have reported that Ras-related C3 botulinum toxin substrate 2 (Rac2), a regulatory subunit of the NADPH oxidase complex, is upregulated in DADS-induced apoptosis in human leukaemia HL-60 cells. The aim of the present study was to investigate the role of Rac2, NADPH oxidase and reactive oxygen species (ROS) in DADS-induced apoptosis. 2. Expression of the Rac2 gene along with that of five other genes of NADPH oxidase subunits were in HL-60 cells measured by Sybergreen quantitative real-time polymerase chain reaction. RNA interference was used to test the effect of Rac2. Protein expression was evaluated using western blot analysis and ROS levels were measured by 2',7'-dichlorofluorescein diacetate (DCFH-DA) fluorescence. DNA fragmentation and flow cytometry analysis were used to detect apoptotic cells. 3. Levels of Rac2 gene and protein were significantly upregulated and NADPH oxidase was activated in DADS-induced apoptosis. Pretreatment of HL-60 cells with small interfering (si) RNAs to inhibit Rac2 blocked DADS-induced apoptosis. Diallyl disulphide-induced intracellular ROS production was increased in phorbol myristate acetate-stimulated cells, but decreased in Rac2 siRNA-treated cells. In Rac2 siRNA-treated cells, activator protein-1 and caspase 3 levels decreased, c-myc protein levels were increased and p38 protein levels were unchanged compared with Rac2-competent, DADS-treated cells. 4. These results demonstrate that NADPH oxidase is the main source of DADS-induced ROS. In addition, Rac2 selectively activates the c-Jun N-terminal kinase pathway, but not the p38 pathway, in DADS-induced apoptosis. So, Rac2, NADPH oxidase and ROS have a critical role in DADS-induced apoptosis in human leukaemia HL-60 cells.
Collapse
Affiliation(s)
- Lan Yi
- Cancer Research Institute, University of South China, Hengyang, Hunan Province, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Association of Toll-like receptor signaling and reactive oxygen species: a potential therapeutic target for posttrauma acute lung injury. Mediators Inflamm 2010; 2010. [PMID: 20706658 PMCID: PMC2913855 DOI: 10.1155/2010/916425] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 06/01/2010] [Accepted: 06/08/2010] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) frequently occurs in traumatic patients and serves as an important component of systemic inflammatory response syndrome (SIRS). Hemorrhagic shock (HS) that results from major trauma promotes the development of SIRS and ALI by priming the innate immune system for an exaggerated inflammatory response. Recent studies have reported that the mechanism underlying the priming of pulmonary inflammation involves the complicated cross-talk between Toll-like receptors (TLRs) and interactions between neutrophils (PMNs) and alveolar macrophages (AMvarphi) as well as endothelial cells (ECs), in which reactive oxygen species (ROS) are the key mediator. This paper summarizes some novel mechanisms underlying HS-primed lung inflammation focusing on the role of TLRs and ROS, and therefore suggests a new therapeutic target for posttrauma ALI.
Collapse
|
37
|
Hwang JJ, Kim HN, Kim J, Cho DH, Kim MJ, Kim YS, Kim Y, Park SJ, Koh JY. Zinc(II) ion mediates tamoxifen-induced autophagy and cell death in MCF-7 breast cancer cell line. Biometals 2010; 23:997-1013. [DOI: 10.1007/s10534-010-9346-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 05/12/2010] [Indexed: 12/25/2022]
|
38
|
Tostmann A, Aarnoutse RE, Peters WH, Richard PR, Boeree MJ. Xanthine oxidase inhibition by allopurinol increasesin vitropyrazinamide-induced hepatotoxicity in HepG2 cells. Drug Chem Toxicol 2010; 33:325-8. [DOI: 10.3109/01480540903449715] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Involvement of mTOR and survivin inhibition in tamoxifen-induced apoptosis in human hepatoblastoma cell line HepG2. Biomed Pharmacother 2010; 64:249-53. [DOI: 10.1016/j.biopha.2009.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 06/07/2009] [Indexed: 12/21/2022] Open
|
40
|
c-Jun activation is required for 4-hydroxytamoxifen-induced cell death in breast cancer cells. Oncogene 2009; 29:978-91. [DOI: 10.1038/onc.2009.400] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Tamoxifen inhibits proliferation and induces apoptosis in human hepatocellular carcinoma cell line HepG2 via down-regulation of survivin expression. Biomed Pharmacother 2009; 63:375-9. [DOI: 10.1016/j.biopha.2008.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 09/26/2008] [Indexed: 10/21/2022] Open
|
42
|
Payré B, de Medina P, Boubekeur N, Mhamdi L, Bertrand-Michel J, Tercé F, Fourquaux I, Goudounèche D, Record M, Poirot M, Silvente-Poirot S. Microsomal antiestrogen-binding site ligands induce growth control and differentiation of human breast cancer cells through the modulation of cholesterol metabolism. Mol Cancer Ther 2009; 7:3707-18. [PMID: 19074846 DOI: 10.1158/1535-7163.mct-08-0507] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The microsomal antiestrogen-binding site (AEBS) is a high-affinity membranous binding site for the antitumor drug tamoxifen that selectively binds diphenylmethane derivatives of tamoxifen such as PBPE and mediates their antiproliferative properties. The AEBS is a hetero-oligomeric complex consisting of 3beta-hydroxysterol-Delta8-Delta7-isomerase and 3beta-hydroxysterol-Delta7-reductase. High-affinity AEBS ligands inhibit these enzymes leading to the massive intracellular accumulation of zymostenol or 7-dehydrocholesterol (DHC), thus linking AEBS binding to the modulation of cholesterol metabolism and growth control. The aim of the present study was to gain more insight into the control of breast cancer cell growth by AEBS ligands. We report that PBPE and tamoxifen treatment induced differentiation in human breast adenocarcinoma cells MCF-7 as indicated by the arrest of cells in the G0-G1 phase of the cell cycle, the increase in the cell volume, the accumulation and secretion of lipids, and a milk fat globule protein found in milk. These effects were observed with other AEBS ligands and with zymostenol and DHC. Vitamin E abrogates the induction of differentiation and reverses the control of cell growth produced by AEBS ligands, zymostenol, and DHC, showing the importance of the oxidative processes in this effect. AEBS ligands induced differentiation in estrogen receptor-negative mammary tumor cell lines SKBr-3 and MDA-MB-468 but with a lower efficiency than observed with MCF-7. Together, these data show that AEBS ligands exert an antiproliferative effect on mammary cancer cells by inducing cell differentiation and growth arrest and highlight the importance of cholesterol metabolism in these effects.
Collapse
Affiliation(s)
- Bruno Payré
- INSERM, U-563, Section métabolisme, oncogenèse et différenciation cellulaire, Centre de Physiopathologie de Toulouse Purpan, Institut Claudius Regaud, 24 rue du Pont Saint Pierre, F-31052 Toulouse Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kadara H, Tahara E, Kim HJ, Lotan D, Myers J, Lotan R. Involvement of Rac in fenretinide-induced apoptosis. Cancer Res 2008; 68:4416-23. [PMID: 18519704 DOI: 10.1158/0008-5472.can-08-0031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) has shown potential as a chemopreventive and therapeutic agent. The ability of 4HPR to enhance production of reactive oxygen species (ROS) leading to apoptosis has been suggested as a possible mechanism underlying these effects. We explored the possibility that ROS induction by 4HPR involves the small GTPase Ras-related C3 botulinum toxin substrate (Rac), a regulatory subunit of the NADPH oxidase complex. Rac was activated in human head and neck squamous cell carcinoma (HNSCC) cells as early as 5 minutes following 4HPR exposure. Moreover, inhibition of Rac activity or silencing of its expression by RNA interference decreased ROS generation in human head and neck, lung, and cervical cancer cells and murine melanoma cells. In HNSCC UMSCC-22B cells, this decrease correlated with reduction in apoptosis induction by 4HPR. Expression of a constitutive active mutant Rac increased basal and 4HPR-induced ROS generation and poly(ADP-ribose) polymerase cleavage. In addition, the metastatic DM14 cells exhibited higher Rac activation following 4HPR treatment compared with the primary Tu167-C2 cells. Furthermore, the metastatic cancer cells tested exhibited higher ROS generation and growth inhibition due to 4HPR exposure compared with their primary cancer cell counterparts. These findings show a preferential susceptibility of metastatic cells to the proapoptotic retinoid 4HPR through Rac activation and support the use of ROS-inducing agents such as 4HPR against metastatic cancer cells.
Collapse
Affiliation(s)
- Humam Kadara
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
44
|
Nong Q, Komatsu M, Izumo K, Indo HP, Xu B, Aoyama K, Majima HJ, Horiuchi M, Morimoto K, Takeuchi T. Involvement of reactive oxygen species in Microcystin-LR-induced cytogenotoxicity. Free Radic Res 2008; 41:1326-37. [PMID: 17963120 DOI: 10.1080/10715760701704599] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Microcystin-LR (MCLR) is a potent hepatotoxin. Oxidative stress is thought to be implicated in the cytotoxicity of MCLR, but the mechanisms by which MCLR produces reactive oxygen species (ROS) are still unclear. This study investigated the role and possible sources of ROS generation in MCLR-induced cytogenotoxicity in HepG2, a human hepatoma cell line. MCLR increased DNA strand breaks, 8-hydroxydeoxiguanosine formation, lipid peroxidation, as well as LDH release, all of which were inhibited by ROS scavengers. ROS scavengers partly suppressed MCLR-induced cytotoxicity determined by the MTT assay. MCLR induced the generation of ROS, as confirmed by confocal microscopy with 2-[6-(4'-hydroxy)phenoxy-3H-xanthen-3-on-9-yl]benzoic acid, and upregulated the expression of CYP2E1 mRNA. In addition, CYP2E1 inhibitors chlormethiazole and diallyl dulphide inhibited both ROS generation and cytotoxicity induced by MCLR. The results suggest that ROS contribute to MCLR-induced cytogenotoxicity. CYP2E1 might be a potential source responsible for ROS generation by MCLR.
Collapse
Affiliation(s)
- Qingqing Nong
- Department of Environmental Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sharma R, Traore K, Trush MA, Rose NR, Burek CL. Intracellular adhesion molecule-1 up-regulation on thyrocytes by iodine of non-obese diabetic.H2(h4) mice is reactive oxygen species-dependent. Clin Exp Immunol 2008; 152:13-20. [PMID: 18241232 DOI: 10.1111/j.1365-2249.2008.03590.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Intracellular adhesion molecule-1 (ICAM-1) expression on the thyroid follicular cells of non-obese diabetic (NOD).H2(h4) mice is enhanced by iodide treatment, which correlates with autoimmune thyroid disease in genetically susceptible NOD.H2(h4) mice. The current study examines the mechanism of iodine-enhanced up-regulation of ICAM-1 on the surface of thyroid cells. We hypothesized that the up-regulation of ICAM-1 is due to a transient increase in production of reactive oxygen species (ROS). ROS may initiate signalling of the ICAM-1 gene promoter, enhancing up-regulated ICAM-1 protein on the cell surface. Single-cell suspensions of thyroid follicular cells from thyroiditis-susceptible NOD.H2(h4) or non-susceptible BALB/c mice were treated in vitro with sodium iodide. Extracellular and intracellular ROS were assessed by luminol-derived chemiluminescence and flow cytometry assays respectively. Our results demonstrate that thyroid follicular cells of NOD.H2(h4) generate higher levels of ROS compared with cells from non-susceptible strains of mice. Expression of a subunit protein of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, p67(phox), was analysed by Western blot immunoassay. A constitutive expression of the p67(phox) subunit protein was observed in NOD.H2(h4) mice prior to iodine treatment. No such expression was found in BALB/c mice. Treatment of NOD.H2(h4) thyroid cells with diphenyleneiodium, an inhibitor of NADPH oxidase, reduced generation of ROS and of ICAM-1 protein expression. Thus, thyrocytes from NOD.H2(h4) mice produce enhanced levels of ROS that may be mediated by NADPH oxidase. Consequently, in NOD.H2(h4) mice the ROS-induced signal for ICAM-1 up-regulation may contribute to mononuclear cellular infiltration of the thyroid gland and the progression of autoimmune thyroid disease.
Collapse
Affiliation(s)
- R Sharma
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
46
|
Choi SI, Jeong CS, Cho SY, Lee YS. Mechanism of apoptosis induced by apigenin in HepG2 human hepatoma cells: involvement of reactive oxygen species generated by NADPH oxidase. Arch Pharm Res 2007; 30:1328-35. [PMID: 18038912 DOI: 10.1007/bf02980274] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Although plant-derived flavonoids have been reported to have anti-cancer activities, the exact mechanism of these actions is not completely understood. In this study we investigated the role for reactive oxygen species (ROS) as a mediator of the apoptosis induced by apigenin, a widespread flavonoid in plant, in HepG2 human hepatoma cells. Apigenin reduced cell viability, and induced apoptotic cell death in a dose-dependent manner. In addition, it evoked a dose-related elevation of intracellular ROS level. Treatment with various inhibitors of the NADPH oxidase (diphenylene iodonium, apocynin, neopterine) significantly blunted both the generation of ROS and induction of apoptosis induced by apigenin. These results suggest that ROS generated through the activation of the NADPH oxidase may play an essential role in the apoptosis induced by apigenin in HepG2 cells. These results further suggest that apigenin may be valuable for the therapeutic management of human hepatomas.
Collapse
Affiliation(s)
- Soo Im Choi
- Plant Resources Research Institute, Duksung Women's University, Seoul 132-714, Korea
| | | | | | | |
Collapse
|
47
|
Tabassum H, Parvez S, Rehman H, Banerjee BD, Raisuddin S. Catechin as an antioxidant in liver mitochondrial toxicity: Inhibition of tamoxifen-induced protein oxidation and lipid peroxidation. J Biochem Mol Toxicol 2007; 21:110-7. [PMID: 17623883 DOI: 10.1002/jbt.20167] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tamoxifen (TAM) is a nonsteroidal triphenylethylene antiestrogenic drug widely used in the treatment and prevention of breast cancer. TAM brings about a collapse of the mitochondrial membrane potential. It acts both as an uncoupling agent and as a powerful inhibitor of mitochondrial electron transport chain. The effect of catechin pretreatment on the mitochondrial toxicity of TAM was studied in liver mitochondria of Swiss albino mice. TAM treatment caused a significant increase in the mitochondrial lipid peroxidation (LPO) and the protein carbonyls (PCs). It also caused a significant increase in superoxide radical production. Pretreatment of mice with catechin (40 mg/kg) showed significant protection as demonstrated by marked attenuation of increased oxidative stress parameters such LPO, PCs, and superoxide production. It also restored the decreased nonenzymatic and enzymatic antioxidants of mitochondria. The inhibitory effect of catechin on TAM-:induced oxidative damage suggests that it may have potential benefits in prevention of human diseases where reactive oxygen species have some role as causative agents.
Collapse
Affiliation(s)
- Heena Tabassum
- Immunotoxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110 062, India
| | | | | | | | | |
Collapse
|
48
|
Jatobá CAN, de Rezende AA, de Paiva Rodrigues SJ, de Almeida Câmara MM, das Graças Almeida M, Freire-Neto F, da Rocha LRM, da Medeiros AC, Brandão-Neto J, de Carvalho Formiga MC, de Azevedo IM, de Oliveira Ramos AM. Liver iron overload induced by tamoxifen in diabetic and non-diabetic female Wistar rats. Biometals 2007; 21:171-8. [PMID: 17636394 DOI: 10.1007/s10534-007-9105-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 06/25/2007] [Indexed: 01/11/2023]
Abstract
Tamoxifen (TX), a drug used in the treatment of breast cancer, may cause hepatic changes in some patients. The consequences of its use on the liver tissues of rats with or without diabetes mellitus (DM) have not been fully explored. The purpose of this study was to evaluate the correlation between plasma hepatic enzyme levels and the presence of iron overload in the hepatic tissue of female Wistar rats with or without streptozotocin-induced DM and using TX. Female rats were studied in control groups: C-0 (non-drug users), C-V (sorbitol vehicle only) and C-TX (using TX). DM (diabetic non-drug users) and DM-TX (diabetics using TX) were the test groups. Sixty days after induced DM, blood samples were collected for glucose, alanine aminotransferase (ALT), aspartate aminotransferase (AST) alkaline phosphatase (ALP) and bilirubin measures. Hepatic fragments were processed and stained with hematoxylin and eosin, Masson's trichrome, Perls. The hepatic iron content was quantified by atomic absorption spectrometry. AST, ALT and ALP levels were significantly elevated in the DM and DM-TX groups, with unchanged bilirubin levels. Liver iron overload using Perls stain and atomic absorption spectrometry were observed exclusively in groups C-TX and DM-TX. There was positive correlation between AST, ALT and ALP levels and microscopic hepatic siderosis intensity in group DM-TX. In conclusion, TX administration is associated with liver siderosis in diabetic and non-diabetic rats. In addition, TX induced liver iron overload with unaltered hepatic function in non-diabetic rats and may be a useful tool for investigating the biological control of iron metabolism.
Collapse
Affiliation(s)
- Carlos André Nunes Jatobá
- Postgraduate Course in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
López-Lázaro M. Dual role of hydrogen peroxide in cancer: Possible relevance to cancer chemoprevention and therapy. Cancer Lett 2007; 252:1-8. [PMID: 17150302 DOI: 10.1016/j.canlet.2006.10.029] [Citation(s) in RCA: 498] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 10/30/2006] [Accepted: 10/31/2006] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that hydrogen peroxide (H(2)O(2)) plays an important role in cancer development. Experimental data have shown that cancer cells produce high amounts of H(2)O(2). An increase in the cellular levels of H(2)O(2) has been linked to several key alterations in cancer, including DNA alterations, cell proliferation, apoptosis resistance, metastasis, angiogenesis and hypoxia-inducible factor 1 (HIF-1) activation. It has also been observed that the malignant phenotype of cancer cells can be reversed just by decreasing the cellular levels of H(2)O(2). On the other hand, there is evidence that H(2)O(2) can induce apoptosis in cancer cells selectively and that the activity of several anticancer drugs commonly used in the clinic is mediated, at least in part, by H(2)O(2). The present report discusses that the high levels of H(2)O(2) commonly observed in cancer cells may be essential for cancer development; these high levels, however, seem almost incompatible with cell survival and may make cancer cells more susceptible to H(2)O(2)-induced cell death than normal cells. An understanding of this dual role of H(2)O(2) in cancer might be exploited for the development of cancer chemopreventive and therapeutic strategies.
Collapse
Affiliation(s)
- Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Spain.
| |
Collapse
|
50
|
Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007; 87:245-313. [PMID: 17237347 DOI: 10.1152/physrev.00044.2005] [Citation(s) in RCA: 5065] [Impact Index Per Article: 281.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
For a long time, superoxide generation by an NADPH oxidase was considered as an oddity only found in professional phagocytes. Over the last years, six homologs of the cytochrome subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the phagocyte NADPH oxidase itself (NOX2/gp91(phox)), the homologs are now referred to as the NOX family of NADPH oxidases. These enzymes share the capacity to transport electrons across the plasma membrane and to generate superoxide and other downstream reactive oxygen species (ROS). Activation mechanisms and tissue distribution of the different members of the family are markedly different. The physiological functions of NOX family enzymes include host defense, posttranlational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. NOX enzymes also contribute to a wide range of pathological processes. NOX deficiency may lead to immunosuppresion, lack of otoconogenesis, or hypothyroidism. Increased NOX activity also contributes to a large number or pathologies, in particular cardiovascular diseases and neurodegeneration. This review summarizes the current state of knowledge of the functions of NOX enzymes in physiology and pathology.
Collapse
Affiliation(s)
- Karen Bedard
- Biology of Ageing Laboratories, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|