1
|
Munteanu A, Gogulescu A, Șoica C, Mioc A, Mioc M, Milan A, Lukinich-Gruia AT, Pricop MA, Jianu C, Banciu C, Racoviceanu R. In Vitro and In Silico Evaluation of Syzygium aromaticum Essential Oil: Effects on Mitochondrial Function and Cytotoxic Potential Against Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2024; 13:3443. [PMID: 39683236 DOI: 10.3390/plants13233443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The current study proposes the in vitro and in silico anticancer evaluation of clove (Syzygium aromaticum L.) essential oil (CEO). The steam hydrodistillation method used yielded 10.7% (wt) CEO. GC-MS analysis revealed that the obtained oil is rich in eugenol (75%), β-caryophyllene (20%), and α- caryophyllene (2.8%) and also contains several other minor components accounting for approximately 1.5%. The DPPH-based scavenging antioxidant activity was assessed for the obtained CEO, exhibiting an IC50 value of 158 μg/mL. The cytotoxic effects of CEO, its major component eugenol, and CEO solubilized with Tween-20 and PEG-400 were tested against both noncancerous HaCaT cells and HT-29 human colorectal adenocarcinoma, RPMI-7951 melanoma, A431 skin carcinoma, and NCI-H460 non-small lung cancer cells, using the Alamar Blue and LDH assay after 48 h treatment. The Tween-20 and PEG-400 CEO formulations, at 200 μg/mL, recorded the highest cytotoxic and selective effects against RPMI-7951 (72.75% and 71.56%), HT-29 (71.51% and 45.43%), and A431 cells (61.62% and 59.65%). Furthermore, CEO disrupted mitochondrial function and uncoupled oxidative phosphorylation. This effect was more potent for the CEO against the RPMI-7951 and HT-29 cells, whereas for the other two tested cell lines, a more potent inhibition of mitochondrial function was attributed to eugenol. The present study is the first to specifically investigate the effects of CEO and Tween-20 and PEG-400 CEO formulations on the mitochondrial function of RPMI-7951, HT-29, A431, and NCI-H460 cancer cell lines using high-resolution respirometry, providing novel insights into their impact on mitochondrial respiration and bioenergetics in cancer cells. The results obtained may explain the increased ROS production observed in cancer cell lines treated with eugenol and CEO. Molecular docking identified potential protein targets, related to the CEO anticancer activity, in the form of PI3Kα, where the highest active theoretical inhibitor was calamenene (-7.5 kcal/mol). Docking results also showed that calamenene was the overall most active theoretical inhibitor for all docked proteins and indicated a potential presence of synergistic effects among all CEO constituents.
Collapse
Affiliation(s)
- Andreea Munteanu
- Department of Internal Medicine IV, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Armand Gogulescu
- Department XVI: Balneology, Medical Rehabilitation and Rheumatology, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Codruța Șoica
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Alexandra Mioc
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Marius Mioc
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Andreea Milan
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | | | - Maria-Alexandra Pricop
- OncoGen Centre, Clinical County Hospital "Pius Branzeu", Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania
- Department of Applied Chemistry and Environmental Engineering and Inorganic Compounds, Faculty of Industrial Chemistry, Biotechnology and Environmental Engineering, Polytechnic University of Timisoara, Vasile Pârvan 6, 300223 Timisoara, Romania
| | - Calin Jianu
- Faculty of Food Engineering, Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania"Timisoara, Calea Aradului 119, 300645 Timisoara, Romania
| | - Christian Banciu
- Department of Internal Medicine IV, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Roxana Racoviceanu
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| |
Collapse
|
2
|
Schmitt L, Krings KS, Wolsing A, Buque X, Zimmermann M, Flores-Romero H, Lenz T, Lechtenberg I, Peter C, Stork B, Teusch N, Proksch P, Stühler K, García-Sáez AJ, Reichert AS, Aspichueta P, Bhatia S, Wesselborg S. Targeting mitochondrial metabolism by the mitotoxin bromoxib in leukemia and lymphoma cells. Cell Commun Signal 2024; 22:541. [PMID: 39533399 PMCID: PMC11558866 DOI: 10.1186/s12964-024-01913-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Targeting mitochondrial metabolism represents a promising approach for cancer treatment. Here, we investigated the mitotoxic potential of the polybrominated diphenyl ether bromoxib, a natural compound isolated from the marine sponge Dysidea family. We could show that bromoxib comprised strong cytotoxicity in different leukemia and lymphoma cell lines (such as HL60, HPBALL, Jurkat, K562, KOPTK1, MOLT4, SUPB15 and Ramos), but also in solid tumor cell lines (such as glioblastoma cell lines SJ-GBM2 and TP365MG). Bromoxib activated the mitochondrial death pathway as evidenced by the rapid translocation of Bax to the mitochondria and the subsequent mitochondrial release of Smac. Accordingly, bromoxib-induced apoptosis was blocked in caspase 9 deficient Jurkat cells and Jurkat cells overexpressing the antiapoptotic protein Bcl-2. In addition, we could show that bromoxib functioned as an uncoupler of the electron transport chain with similar rapid kinetics as CCCP in terms of dissipation of the mitochondrial membrane potential (ΔΨm), processing of the dynamin-like GTPase OPA1 and subsequent fragmentation of mitochondria. Beyond that, bromoxib strongly abrogated ATP production via glycolysis as well as oxidative phosphorylation (OXPHOS) by targeting electron transport chain complexes II, III, and V (ATP-synthase) in Ramos lymphoma cells. Thus, bromoxib's potential to act on both cytosolic glycolysis and mitochondrial respiration renders it a promising agent for the treatment of leukemia and lymphoma.
Collapse
Affiliation(s)
- Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Karina S Krings
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Andre Wolsing
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Xabier Buque
- Department of Physiology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vitoria-gasteiz, Spain
| | - Marcel Zimmermann
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Hector Flores-Romero
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Thomas Lenz
- Molecular Proteomics Laboratory, Biological-Medical-Research Centre (BMFZ), Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Christoph Peter
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Björn Stork
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Nicole Teusch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biological-Medical-Research Centre (BMFZ), Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ana J García-Sáez
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vitoria-gasteiz, Spain
- Biobizkia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
3
|
Wiggins R, Woo J, Mito S. Optimizing Niclosamide for Cancer Therapy: Improving Bioavailability via Structural Modification and Nanotechnology. Cancers (Basel) 2024; 16:3548. [PMID: 39456642 PMCID: PMC11506536 DOI: 10.3390/cancers16203548] [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: 09/15/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Inhibition of multiple cancer-related pathways has made niclosamide a promising candidate for the treatment of various cancers. However, its clinical application has been significantly limited by poor bioavailability. This review will discuss current findings on improving niclosamide bioavailability through modification of its chemical structure and utilization of novel nanotechnologies, like electrospraying and supercritical fluids, to improve drug delivery. For example, niclosamide derivatives, such as o-alkylamino-tethered niclosamide derivates, niclosamide ethanolamine salt, and niclosamide piperazine salt, have demonstrated increased water solubility without compromising anticancer activity in vitro. Additionally, this review briefly discusses recent findings on the first pass metabolism of niclosamide in vivo, the role of cytochrome P450-mediated hydroxylation, UDP-glucuronosyltransferase mediated glucuronidation, and how enzymatic inhibition could enhance niclosamide bioavailability. Ultimately, there is a need for researchers to synthesize, evaluate, and improve upon niclosamide derivatives while experimenting with the employment of nanotechnologies, such as targeted delivery and nanoparticle modification, as a way to improve drug administration. Researchers should strive to improve drug-target accuracy, its therapeutic index, and increase the drug's efficacy as an anti-neoplastic agent.
Collapse
Affiliation(s)
| | | | - Shizue Mito
- Department of Medical Education, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA; (R.W.); (J.W.)
| |
Collapse
|
4
|
Needham D. Niclosamide: A career builder. J Control Release 2024; 369:786-856. [PMID: 37544514 DOI: 10.1016/j.jconrel.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 08/08/2023]
Abstract
My contribution to honoring Professor Kinam Park celebrates and resonates with his scholarly career in drug delivery, his commitment to encouraging the next generation(s), and his efforts to keep us focused on clinically effective formulations. To do this I take as my example, niclosamide, a small molecule protonophore that, uniquely, can "target" all cell membranes, both plasma and organelle. As such, it acts upstream of many cell pathways and so has the potential to affect many of the essential events that a cell, and particularly a diseased cell or other entities like a virus, use to stay alive and prosper. Literature shows that it has so far been discovered to positively influence (at least): cancer, bacterial and viral infection, metabolic diseases such as Type II diabetes, NASH and NAFLD, artery constriction, endometriosis, neuropathic pain, rheumatoid arthritis, sclerodermatous graft-versus-host disease, systemic sclerosis, Parkinson's, and COPD. With such a fundamental action and broad-spectrum activity, I believe that studying niclosamide in all its manifestations, discovering if and to what extent it can contribute positively to disease control (and also where it can't), formulating it as effective therapeutics, and testing them in preclinical and clinical trials is a career builder for our next generation(s). The article is divided into two parts: Part I introduces niclosamide and other proton shunts mainly in cancer and viral infections and reviews an exponentially growing literature with some concepts and physicochemical properties that lead to its proton shunt mechanism. Part II focuses on repurposing by reformulation of niclosamide. I give two examples of "carrier-free formulations", - one for cancer (as a prodrug therapeutic of niclosamide stearate for i.v. and other administration routes, exemplified by our recent work on Osteosarcoma in mice and canine patients), and the other as a niclosamide solution formulation (that could provide the basis for a preventative nasal spray and early treatment option for COVID19 and other respiratory virus infections). My goal is to excite and enthuse, encourage, and motivate all involved in the drug development and testing process in academia, institutes, and industry, to learn more about this interesting molecule and others like it. To enable such endeavors, I give many proposed ideas throughout the document, that have been stimulated and inspired by gaps in the literature, urgent needs in disease, and new studies arising from our own work. The hope is that, by reading through this document and studying the suggested topics and references, the drug delivery and development community will continue our lineage and benefit from our legacy to achieve niclosamide's potential as an effective contributor to the treatment and control of many diseases and conditions.
Collapse
Affiliation(s)
- David Needham
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA; Translational Therapeutics, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
5
|
Mlejnek P. Direct Interaction between N-Acetylcysteine and Cytotoxic Electrophile—An Overlooked In Vitro Mechanism of Protection. Antioxidants (Basel) 2022; 11:antiox11081485. [PMID: 36009205 PMCID: PMC9405167 DOI: 10.3390/antiox11081485] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
In laboratory experiments, many electrophilic cytotoxic agents induce cell death accompanied by reactive oxygen species (ROS) production and/or by glutathione (GSH) depletion. Not surprisingly, millimolar concentrations of N-acetylcysteine (NAC), which is used as a universal ROS scavenger and precursor of GSH biosynthesis, inhibit ROS production, restore GSH levels, and prevent cell death. The protective effect of NAC is generally used as corroborative evidence that cell death induced by a studied cytotoxic agent is mediated by an oxidative stress-related mechanism. However, any simple interpretation of the results of the protective effects of NAC may be misleading because it is unable to interact with superoxide (O2•−), the most important biologically relevant ROS, and is a very weak scavenger of H2O2. In addition, NAC is used in concentrations that are unnecessarily high to stimulate GSH synthesis. Unfortunately, the possibility that NAC as a nucleophile can directly interact with cytotoxic electrophiles to form non-cytotoxic NAC–electrophile adduct is rarely considered, although it is a well-known protective mechanism that is much more common than expected. Overall, apropos the possible mechanism of the cytoprotective effect of NAC in vitro, it is appropriate to investigate whether there is a direct interaction between NAC and the cytotoxic electrophile to form a non-cytotoxic NAC–electrophilic adduct(s).
Collapse
Affiliation(s)
- Petr Mlejnek
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 77515 Olomouc, Czech Republic
| |
Collapse
|
6
|
Shrestha R, Johnson E, Byrne FL. Exploring the therapeutic potential of mitochondrial uncouplers in cancer. Mol Metab 2021; 51:101222. [PMID: 33781939 PMCID: PMC8129951 DOI: 10.1016/j.molmet.2021.101222] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mitochondrial uncouplers are well-known for their ability to treat a myriad of metabolic diseases, including obesity and fatty liver diseases. However, for many years now, mitochondrial uncouplers have also been evaluated in diverse models of cancer in vitro and in vivo. Furthermore, some mitochondrial uncouplers are now in clinical trials for cancer, although none have yet been approved for the treatment of cancer. SCOPE OF REVIEW In this review we summarise published studies in which mitochondrial uncouplers have been investigated as an anti-cancer therapy in preclinical models. In many cases, mitochondrial uncouplers show strong anti-cancer effects both as single agents, and in combination therapies, and some are more toxic to cancer cells than normal cells. Furthermore, the mitochondrial uncoupling mechanism of action in cancer cells has been described in detail, with consistencies and inconsistencies between different structural classes of uncouplers. For example, many mitochondrial uncouplers decrease ATP levels and disrupt key metabolic signalling pathways such as AMPK/mTOR but have different effects on reactive oxygen species (ROS) production. Many of these effects oppose aberrant phenotypes common in cancer cells that ultimately result in cell death. We also highlight several gaps in knowledge that need to be addressed before we have a clear direction and strategy for applying mitochondrial uncouplers as anti-cancer agents. MAJOR CONCLUSIONS There is a large body of evidence supporting the therapeutic use of mitochondrial uncouplers to treat cancer. However, the long-term safety of some uncouplers remains in question and it will be critical to identify which patients and cancer types would benefit most from these agents.
Collapse
Affiliation(s)
- Riya Shrestha
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia
| | - Edward Johnson
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia
| | - Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia.
| |
Collapse
|
7
|
Chow LWC, Cheng KS, Leong F, Cheung CW, Shiao LR, Leung YM, Wong KL. Enhancing tetrandrine cytotoxicity in human lung carcinoma A549 cells by suppressing mitochondrial ATP production. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2019; 392:427-436. [PMID: 30547225 DOI: 10.1007/s00210-018-01601-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022]
Abstract
ATP depletion induced by inhibiting glycolysis or mitochondrial ATP production has been demonstrated to cause cancer cell death. Whether ATP depletion can enhance the efficacy and potency of anti-cancer effects of herbal compounds is so far unknown. We examined the enhancing effect of ATP depletion on anti-cancer actions of tetrandrine (TET) in human lung carcinoma A549 cells. A 24-h incubation of A549 cells with tetrandrine caused a concentration-dependent cytotoxic effect (LC50 = 66.1 μM). Co-incubation with 20 mM 2-deoxyglucose (2-DG, glycolysis inhibitor) caused only a very slight enhancement of tetrandrine cytotoxicity. By contrast, inhibiting mitochondrial ATP production with oligomycin (10 μM, ATP synthase inhibitor) and FCCP (30 μM, uncoupling agent) (thus, oligo-FCCP) on its own caused only slight cell cytotoxicity but strongly potentiated tetrandrine cytotoxicity (tetrandrine LC50 = 15.6 μM). The stronger enhancing effect of oligo-FCCP than 2-DG on TET toxicity did not result from more severe overall ATP depletion, since both treatments caused a similar ATP level suppression. Neither oligo-FCCP nor 2-DG synergized with tetrandrine in decreasing mitochondrial membrane potential. TET on its own triggered reactive oxygen species (ROS) production, and oligo-FCCP, but not 2-DG, potentiated TET in causing ROS production. Taken together, our results suggest that inhibiting ATP production from mitochondria, but not from glycolysis, appears to be a very effective means in augmenting TET-triggered ROS production and hence toxicity in A549 cells.
Collapse
Affiliation(s)
- Louis W C Chow
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- UNIMED Medical Institute and Organisation for Oncology and Translational Research, Hong Kong, China
- Organisation for Oncology and Translational Research, Hong Kong, China
| | - Ka-Shun Cheng
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Fai Leong
- Department of Anaesthesiology of Centro Hospitalar conde de Sao Januario, Macao Health Bureau, Macau, SAR, China
| | - Chi-Wai Cheung
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lian-Ru Shiao
- Department of Physiology, China Medical University, No.91, Hsueh-Shih Road, Taichung, 40402, Taiwan, Republic of China
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, No.91, Hsueh-Shih Road, Taichung, 40402, Taiwan, Republic of China.
| | - Kar-Lok Wong
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan.
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
Park WH. Gallic acid induces HeLa cell death via increasing GSH depletion rather than ROS levels. Oncol Rep 2016; 37:1277-1283. [DOI: 10.3892/or.2016.5335] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/19/2016] [Indexed: 11/05/2022] Open
|
9
|
Satoh K, Zhang L, Zhang Y, Chelluri R, Boufraqech M, Nilubol N, Patel D, Shen M, Kebebew E. Identification of Niclosamide as a Novel Anticancer Agent for Adrenocortical Carcinoma. Clin Cancer Res 2016; 22:3458-66. [PMID: 26873959 DOI: 10.1158/1078-0432.ccr-15-2256] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/05/2016] [Indexed: 01/09/2023]
Abstract
PURPOSE Adrenocortical carcinoma (ACC) is a rare and aggressive cancer, and no current effective therapy is available for locally advanced and metastatic ACC. Drug repurposing is an emerging approach for identifying new indications for existing drugs, especially for rare cancers such as ACC. The objective of this study was to use quantitative high-throughput screening to identify agents with antineoplastic activity against ACC. EXPERIMENTAL DESIGN A screening of 4,292 compounds was performed on three ACC cell lines: BD140A, SW-13, and NCI-H295R. RESULTS Twenty-one active compounds were identified, with an efficacy of >80% in all three cell lines. Of these, niclosamide showed higher efficacy and lower IC50 than established anti-ACC drugs. We then validated niclosamide-inhibited cellular proliferation in all three ACC cell lines. Next, we investigated the mechanism by which niclosamide inhibited ACC cell proliferation, and found that it induced caspase-dependent apoptosis and G1 cell-cycle arrest. Niclosamide also decreased cellular migration and reduced the level of mediators of epithelial-to-mesenchymal transition, such as N-cadherin and vimentin. Furthermore, niclosamide treatment resulted in decreased expression of β-catenin. We also evaluated the effect of niclosamide on energy metabolism in ACC cell lines and found it resulted in mitochondrial uncoupling. Niclosamide treatment inhibited ACC tumor growth with no observed toxicity in mice in vivo CONCLUSIONS Our findings suggest that niclosamide has anti-ACC activity through its inhibition of multiple altered cellular pathways and cellular metabolism in ACC. Our results provide a preclinical rationale for evaluating niclosamide therapy in a clinical trial for ACC. Clin Cancer Res; 22(14); 3458-66. ©2016 AACR.
Collapse
Affiliation(s)
- Kei Satoh
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland. Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yaqin Zhang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Raju Chelluri
- SUNY Upstate Medical University at Syracuse, Syracuse, New York
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Naris Nilubol
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Dhaval Patel
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Min Shen
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
10
|
Figarola JL, Singhal J, Tompkins JD, Rogers GW, Warden C, Horne D, Riggs AD, Awasthi S, Singhal SS. SR4 Uncouples Mitochondrial Oxidative Phosphorylation, Modulates AMP-dependent Kinase (AMPK)-Mammalian Target of Rapamycin (mTOR) Signaling, and Inhibits Proliferation of HepG2 Hepatocarcinoma Cells. J Biol Chem 2015; 290:30321-41. [PMID: 26534958 DOI: 10.1074/jbc.m115.686352] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Indexed: 01/01/2023] Open
Abstract
Mitochondrial oxidative phosphorylation produces most of the energy in aerobic cells by coupling respiration to the production of ATP. Mitochondrial uncouplers, which reduce the proton gradient across the mitochondrial inner membrane, create a futile cycle of nutrient oxidation without generating ATP. Regulation of mitochondrial dysfunction and associated cellular bioenergetics has been recently identified as a promising target for anticancer therapy. Here, we show that SR4 is a novel mitochondrial uncoupler that causes dose-dependent increase in mitochondrial respiration and dissipation of mitochondrial membrane potential in HepG2 hepatocarcinoma cells. These effects were reversed by the recoupling agent 6-ketocholestanol but not cyclosporin A and were nonexistent in mitochondrial DNA-depleted HepG2 cells. In isolated mouse liver mitochondria, SR4 similarly increased oxygen consumption independent of adenine nucleotide translocase and uncoupling proteins, decreased mitochondrial membrane potential, and promoted swelling of valinomycin-treated mitochondria in potassium acetate medium. Mitochondrial uncoupling in HepG2 cells by SR4 results in the reduction of cellular ATP production, increased ROS production, activation of the energy-sensing enzyme AMPK, and inhibition of acetyl-CoA carboxylase and mammalian target of rapamycin signaling pathways, leading to cell cycle arrest and apoptosis. Global analysis of SR4-associated differential gene expression confirms these observations, including significant induction of apoptotic genes and down-regulation of cell cycle, mitochondrial, and oxidative phosphorylation pathway transcripts at 24 h post-treatment. Collectively, our studies demonstrate that the previously reported indirect activation of AMPK and in vitro anticancer properties of SR4 as well as its beneficial effects in both animal xenograft and obese mice models could be a direct consequence of its mitochondrial uncoupling activity.
Collapse
Affiliation(s)
- James L Figarola
- From the Departments of Diabetes and Metabolic Diseases Research
| | - Jyotsana Singhal
- From the Departments of Diabetes and Metabolic Diseases Research
| | | | - George W Rogers
- Seahorse Biosciences, North Billerica, Massachusetts 01862, and
| | - Charles Warden
- the Bioinformatics Program, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Arthur D Riggs
- From the Departments of Diabetes and Metabolic Diseases Research
| | - Sanjay Awasthi
- Medical Oncology, Beckman Research Institute of the City of Hope, Comprehensive Cancer Center, Duarte, California 91010
| | - Sharad S Singhal
- From the Departments of Diabetes and Metabolic Diseases Research,
| |
Collapse
|
11
|
Mlejnek P, Dolezel P. Loss of mitochondrial transmembrane potential and glutathione depletion are not sufficient to account for induction of apoptosis by carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone in human leukemia K562 cells. Chem Biol Interact 2015; 239:100-10. [DOI: 10.1016/j.cbi.2015.06.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 06/07/2015] [Accepted: 06/22/2015] [Indexed: 12/20/2022]
|
12
|
The inhibition of activated hepatic stellate cells proliferation by arctigenin through G0/G1 phase cell cycle arrest: Persistent p27Kip1 induction by interfering with PI3K/Akt/FOXO3a signaling pathway. Eur J Pharmacol 2015; 747:71-87. [DOI: 10.1016/j.ejphar.2014.11.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/06/2014] [Accepted: 11/20/2014] [Indexed: 01/18/2023]
|
13
|
You BR, Kim SH, Park WH. Reactive oxygen species, glutathione, and thioredoxin influence suberoyl bishydroxamic acid-induced apoptosis in A549 lung cancer cells. Tumour Biol 2014; 36:3429-39. [PMID: 25537089 DOI: 10.1007/s13277-014-2978-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/11/2014] [Indexed: 12/12/2022] Open
Abstract
Suberoyl bishydroxamic acid (SBHA) as a histone deacetylase (HDAC) inhibitor can induce apoptosis through the formation of reactive oxygen species (ROS). However, there is no report about the regulation of ROS and antioxidant enzymes in SBHA-treated lung cancer cells. Here, we investigated the toxicological effects of SBHA on the regulations of ROS, glutathione (GSH), and antioxidant enzymes, especially thioredoxin (Trx) in A549 lung cancer cells. SBHA inhibited the growth of A549 cells in time- and dose-dependent manners, and it induced apoptosis which accompanied by the loss of mitochondrial membrane potential (MMP; ΔΨm). SBHA significantly increased ROS levels including O2 (•-) level at 72 h whereas it decreased ROS levels at the early time points (30 min to 3 h). SBHA also induced GSH depletion at 24 and 72 h. N-acetyl cysteine (NAC; a well-known antioxidant) prevented apoptotic cell death and GSH depletion via decreasing ROS in SBHA-treated A549 cells. In addition, SBHA changed the levels of antioxidant-related proteins, especially Trx1. The expression and activity of Trx1 in A549 cells were reduced by SBHA. While the downregulation of Trx1 enhanced cell death, ROS level, and GSH depletion in SBHA-treated A549 cells, the overexpression of Trx1 decreased ROS level in these cells without the prevention of cell death and GSH depletion. In conclusion, SBHA-induced A549 cell death was influenced by changes in ROS and GSH levels. The basal status of Trx1 among other antioxidant proteins was closely correlated with the survival of A549 cells.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, JeonJu, 561-180, Republic of Korea
| | | | | |
Collapse
|
14
|
Tiribuzi R, Tartacca F, Aisa MC, Cerulli GG, Palmerini CA. The impact of nitric oxide on calcium homeostasis in PE/CA-PJ15 cells. Arch Oral Biol 2014; 59:1377-83. [DOI: 10.1016/j.archoralbio.2014.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 11/27/2022]
|
15
|
PX-12 induces apoptosis in Calu-6 cells in an oxidative stress-dependent manner. Tumour Biol 2014; 36:2087-95. [PMID: 25391429 DOI: 10.1007/s13277-014-2816-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/04/2014] [Indexed: 02/06/2023] Open
Abstract
PX-12 (1-methylpropyl 2-imidazolyl disulfide) as a thioredoxin (Trx) inhibitor has an anti-tumor effect. However, there is no report about the toxicological effect of PX-12 on lung cancer cells. Here, we investigated the anti-growth effects of PX-12 on Calu-6 lung cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. PX-12 induced the growth inhibition of Calu-6 cells with IC50 of nearly 3 μM at 72 h. In contrast, PX-12 did not affect the growth of human small airway epithelial cells (HSAECs). Cell cycle distribution analysis indicated that PX-12 significantly induced a G2/M phase arrest in Calu-6 cells. PX-12 also increased the number of annexin V-FITC-positive cells in Calu-6 cells. All the tested caspase inhibitors markedly prevented Calu-6 cell death induced by PX-12. With regard to ROS and GSH levels, PX-12 increased ROS levels containing O2(·-) in Calu-6 cells and induced the depletion of GSH. N-acetyl cysteine (NAC), which is a well-known antioxidant, significantly reduced O2(·-) level in PX-12-treated Calu-6 cells and prevented apoptosis and GSH depletion in these cells. In conclusion, it is the first report that PX-12 inhibited the growth of Calu-6 cells via a G2/M phase arrest as well as apoptosis, which effect was related to the intracellular increases in ROS levels.
Collapse
|
16
|
YOU BORA, SHIN HYERIM, HAN BORAM, KIM SUHNHEE, PARK WOOHYUN. Auranofin induces apoptosis and necrosis in HeLa cells via oxidative stress and glutathione depletion. Mol Med Rep 2014; 11:1428-34. [DOI: 10.3892/mmr.2014.2830] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 06/24/2014] [Indexed: 11/05/2022] Open
|
17
|
Jara JA, Castro-Castillo V, Saavedra-Olavarría J, Peredo L, Pavanni M, Jaña F, Letelier ME, Parra E, Becker MI, Morello A, Kemmerling U, Maya JD, Ferreira J. Antiproliferative and uncoupling effects of delocalized, lipophilic, cationic gallic acid derivatives on cancer cell lines. Validation in vivo in singenic mice. J Med Chem 2014; 57:2440-54. [PMID: 24568614 DOI: 10.1021/jm500174v] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor cells principally exhibit increased mitochondrial transmembrane potential (ΔΨ(m)) and altered metabolic pathways. The therapeutic targeting and delivery of anticancer drugs to the mitochondria might improve treatment efficacy. Gallic acid exhibits a variety of biological activities, and its ester derivatives can induce mitochondrial dysfunction. Four alkyl gallate triphenylphosphonium lipophilic cations were synthesized, each differing in the size of the linker chain at the cationic moiety. These derivatives were selectively cytotoxic toward tumor cells. The better compound (TPP(+)C10) contained 10 carbon atoms within the linker chain and exhibited an IC50 value of approximately 0.4-1.6 μM for tumor cells and a selectivity index of approximately 17-fold for tumor compared with normal cells. Consequently, its antiproliferative effect was also assessed in vivo. The oxygen consumption rate and NAD(P)H oxidation levels increased in the tumor cell lines (uncoupling effect), resulting in a ΔΨ(m) decrease and a consequent decrease in intracellular ATP levels. Moreover, TPP(+)C10 significantly inhibited the growth of TA3/Ha tumors in mice. According to these results, the antineoplastic activity and safety of TPP(+)C10 warrant further comprehensive evaluation.
Collapse
Affiliation(s)
- José A Jara
- Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile , Independencia 1027, Santiago 8380453, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
YOU BORA, PARK WOOHYUN. Suberoylanilide hydroxamic acid-induced HeLa cell death is closely correlated with oxidative stress and thioredoxin 1 levels. Int J Oncol 2014; 44:1745-55. [PMID: 24626405 DOI: 10.3892/ijo.2014.2337] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/06/2014] [Indexed: 11/05/2022] Open
|
19
|
You BR, Shin HR, Park WH. PX-12 inhibits the growth of A549 lung cancer cells via G2/M phase arrest and ROS-dependent apoptosis. Int J Oncol 2013; 44:301-8. [PMID: 24172913 DOI: 10.3892/ijo.2013.2152] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/01/2013] [Indexed: 11/05/2022] Open
Abstract
PX-12 (1-methylpropyl 2-imidazolyl disulfide) is an inhibitor of thioredoxin (Trx-1), which has antitumor effects. However, little is known about the toxicological effect of PX-12 on cancer cells. We investigated the anti-growth effects of PX-12 on A549 lung cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. Based on MTT assays, PX-12 inhibited the growth of A549 cells with an IC50 of approximately 20 µM at 72 h. DNA flow cytometric analysis indicated that PX-12 significantly induced the G2/M phase arrest of the cell cycle in A549 cells. This agent also induced apoptotic cell death, as demonstrated by Annexin V-FITC staining cells and the loss of mitochondrial membrane potential MMP (∆ψm). In addition, the administration of Bax siRNA attenuated PX-12-induced A549 cell death. All the tested caspase inhibitors, especially Z-VAD significantly prevented apoptosis induced by PX-12. With respect to ROS and GSH levels, PX-12 increased ROS levels including O2(•)- in A549 cells and induced GSH depletion. N-acetyl cysteine (NAC) markedly reduced ROS levels in PX-12-treated A549 cells. NAC also prevented apoptotic cell death and GSH depletion induced by PX-12. This is the first report to show that PX-12 inhibits the growth of A549 cells via G2/M phase arrest, and Bax-mediated and ROS-dependent apoptosis.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju 561-180, Republic of Korea
| | | | | |
Collapse
|
20
|
Shin HR, You BR, Park WH. PX-12-induced HeLa cell death is associated with oxidative stress and GSH depletion. Oncol Lett 2013; 6:1804-1810. [PMID: 24260080 PMCID: PMC3833989 DOI: 10.3892/ol.2013.1637] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023] Open
Abstract
PX-12, as an inhibitor of thioredoxin (Trx), has antitumor activity. However, little is known about the toxicological effect of PX-12 on cervical cancer cells. In the present study, the growth inhibitory effects of PX-12 on HeLa cervical cancer cells in association with reactive oxygen species (ROS) and glutathione (GSH) levels were investigated. Based on MTT assays, PX-12 inhibited the growth of HeLa cells with an IC50 value of ~7 μM at 72 h. DNA flow cytometry analysis indicated that 5 and 10 μM PX-12 significantly induced a G2/M phase arrest of the cell cycle. PX-12 also increased the number of dead cells and annexin V-fluorescein isothiocyanate-positive cells, which was accompanied by the loss of mitochondrial membrane potential. All the investigated caspase inhibitors significantly rescued certain cells from PX-12-induced HeLa cell death. With respect to ROS and GSH levels, PX-12 increased ROS levels (including O2•-) in HeLa cells and induced GSH depletion. N-acetyl cysteine markedly reduced the levels of O2•- in PX-12-treated HeLa cells, and prevented apoptotic cell death and GSH depletion in these cells. By contrast, L-buthionine sulfoximine intensified cell death and GSH depletion in PX-12-treated HeLa cells. To conclude, this is the first study to demonstrate that PX-12 inhibits the growth of HeLa cells via G2/M phase arrest, as well as inhibiting apoptosis; the effect was associated with intracellular increases in ROS levels and GSH depletion.
Collapse
Affiliation(s)
- Hye Rim Shin
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju 561-180, Republic of Korea
| | | | | |
Collapse
|
21
|
Han BR, You BR, Park WH. Valproic acid inhibits the growth of HeLa cervical cancer cells via caspase-dependent apoptosis. Oncol Rep 2013; 30:2999-3005. [PMID: 24064712 DOI: 10.3892/or.2013.2747] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 08/26/2013] [Indexed: 11/05/2022] Open
Abstract
Valproic acid (VPA) as a histone deacetylase (HDAC) inhibitor has an anticancer effect. In the present study, we evaluated the effects of VPA on the growth and death of HeLa cervical cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH). Dose- and time-dependent growth inhibition was observed in HeLa cells with an IC50 of approximately 10 mM at 24 h. DNA flow cytometric analysis indicated that 10 mM VPA induced a G2/M phase arrest of the cell cycle. This agent also induced apoptosis, which was accompanied by the cleavage of PARP, the activation of caspase-3, -8 and -9, and the loss of mitochondrial membrane potential (MMP; ∆Ψm). All the tested caspase inhibitors significantly prevented HeLa apoptotic cell death induced by VPA, whereas TNF-α intensified the apoptotic cell death. With respect to ROS and GSH levels, VPA increased ROS levels and induced GSH depletion. However, N-acetyl cysteine (NAC; an antioxidant) and L-buthionine sulfoximine (BSO; a GSH synthesis inhibitor) did not significantly affect cell death in VPA-treated HeLa cells. In conclusion, VPA inhibits the growth of HeLa cervical cancer cells via caspase-dependent apoptosis and the growth inhibition is independent of ROS and GSH level changes.
Collapse
Affiliation(s)
- Bo Ram Han
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju 561-180, Republic of Korea
| | | | | |
Collapse
|
22
|
Park WH. The effect of MAPK inhibitors and ROS modulators on cell growth and death of H₂O₂-treated HeLa cells. Mol Med Rep 2013; 8:557-64. [PMID: 23799549 DOI: 10.3892/mmr.2013.1551] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/10/2013] [Indexed: 11/06/2022] Open
Abstract
Reactive oxygen species (ROS) influence the signaling of mitogen‑activated protein kinases (MAPKs) involved in cell survival and death. In the present study, the toxicological effect of hydrogen peroxide (H2O2) on HeLa cervical cancer cells was evaluated following treatment with MAPK inhibitors [MAP kinase or ERK kinase (MEK), c‑Jun N‑terminal kinase (JNK) or p38], N‑acetyl cysteine (NAC) and propyl gallate (PG) (well‑known antioxidants), or L‑buthionine sulfoximine [BSO; an inhibitor of glutathione (GSH) synthesis]. Treatment with 100 µM H2O2 inhibited the growth of HeLa cells and induced cell death, which was accompanied by loss of the mitochondrial membrane potential (MMP; ΔΨm). H2O2 did not induce any specific phase arrests of the cell cycle. ROS levels increased, while GSH levels decreased in H2O2‑treated HeLa cells after 1 and 24 h of treatment. The MAPK inhibitors enhanced H2O2‑induced HeLa cell death, while only p38 inhibitor increased ROS levels. Both NAC and PG attenuated H2O2‑induced HeLa cell growth inhibition and death together with the suppression of ROS levels. BSO increased ROS levels in H2O2‑treated HeLa cells without increasing cell death. The levels of MMP (ΔΨm) loss and GSH depletion were not closely associated with the levels of apoptosis in HeLa cells treated with the MAPK inhibitors, NAC, PG or BSO, in the presence of H2O2. In conclusion, H2O2 induced HeLa cell growth inhibition and death. MAPK inhibitors generally enhanced H2O2‑induced HeLa cell death. In particular, p38 inhibitor increased ROS levels in H2O2‑treated HeLa cells, while NAC and PG attenuated H2O2‑induced HeLa cell death by suppressing ROS levels.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, JeonJu 561‑180, Republic of Korea.
| |
Collapse
|
23
|
You BR, Park WH. Zebularine inhibits the growth of A549 lung cancer cells via cell cycle arrest and apoptosis. Mol Carcinog 2013; 53:847-57. [PMID: 23661569 DOI: 10.1002/mc.22042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/04/2013] [Indexed: 01/03/2023]
Abstract
Zebularine (Zeb) is a DNA methyltransferase (DNMT) inhibitor to that has an anti-tumor effect. Here, we evaluated the anti-growth effect of Zeb on A549 lung cancer cells in relation to reactive oxygen species (ROS) levels. Zeb inhibited the growth of A549 cells with an IC50 of approximately 70 µM at 72 h. Cell cycle analysis indicated that Zeb induced an S phase arrest in A549 cells. Zeb also induced A549 cell death, which was accompanied by the loss of mitochondrial membrane potential (MMP; ΔΨm ), Bcl-2 decrease, Bax increase, p53 increase and activation of caspase-3 and -8. In contrast, Zeb mildly inhibited the growth of human pulmonary fibroblast (HPF) normal cells and lead to a G1 phase arrest. Zeb did not induce apoptosis in HPF cells. In relation to ROS level, Zeb increased ROS level in A549 cells and induced glutathione (GSH) depletion. The well-known antioxidant, N-acetyl cysteine (NAC) prevented the death of Zeb-treated A549 cells. Moreover, Zeb increased the level of thioredoxin reductase 1 (TrxR1) in A549 cells. While the overexpression of TrxR1 attenuated death and ROS level in Zeb-treated A549 cells, the downregulation of TrxR1 intensified death and ROS level in these cells. In conclusion, Zeb inhibited the growth of A549 lung cancer cells via cell cycle arrest and apoptosis. The inhibition was influenced by ROS and TrxR1 levels.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | | |
Collapse
|
24
|
Park WH, Kim SH. MAPK inhibitors augment gallic acid-induced A549 lung cancer cell death through the enhancement of glutathione depletion. Oncol Rep 2013; 30:513-9. [PMID: 23660987 DOI: 10.3892/or.2013.2447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/12/2013] [Indexed: 11/06/2022] Open
Abstract
Gallic acid (GA) is involved in various biological processes such as cell growth inhibition and apoptosis through changes in reactive oxygen species (ROS). In the present study, we investigated the effects of MAPK (MEK, JNK or p38) inhibitors on cell death in GA-induced A549 lung cancer cells in relation to ROS and glutathione (GSH). Treatment with 100 µM GA inhibited the growth of A549 cells and induced apoptosis and/or necrosis, which was accompanied by the loss of mitochondrial membrane potential (MMP; ∆Ψ(m)). GA increased ROS levels as well as GSH depletion in A549 cells at 24 h. MEK inhibitor seemed to enhance cell growth inhibition by GA. This inhibitor also increased cell death, MMP (∆Ψ(m)) loss and GSH depletion in GA-treated A549 cells. Both JNK and p38 inhibitors intensified growth inhibition, cell death, MMP (∆Ψ(m)) loss and GSH depletion by GA. However, none of the MAPK inhibitors significantly altered ROS levels in GA-treated A549 cells. In conclusion, MAPK inhibitors enhanced growth inhibition and death in GA-treated A549 cells, which were correlated with GSH depletion rather than ROS levels.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju 561-180, Republic of Korea.
| | | |
Collapse
|
25
|
Urra FA, Martínez-Cifuentes M, Pavani M, Lapier M, Jaña-Prado F, Parra E, Maya JD, Pessoa-Mahana H, Ferreira J, Araya-Maturana R. An ortho-carbonyl substituted hydroquinone derivative is an anticancer agent that acts by inhibiting mitochondrial bioenergetics and by inducing G₂/M-phase arrest in mammary adenocarcinoma TA3. Toxicol Appl Pharmacol 2013; 267:218-27. [PMID: 23333614 DOI: 10.1016/j.taap.2012.12.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 10/23/2012] [Accepted: 12/25/2012] [Indexed: 01/16/2023]
Abstract
Tumor cells present a known metabolic reprogramming, which makes them more susceptible for a selective cellular death by modifying its mitochondrial bioenergetics. Anticancer action of the antioxidant 9,10-dihydroxy-4,4-dimethyl-5,8-dihydroanthracen-1(4H)-one (HQ) on mouse mammary adenocarcinoma TA3, and its multiresistant variant TA3-MTXR, were evaluated. HQ decreased the viability of both tumor cells, affecting slightly mammary epithelial cells. This hydroquinone blocked the electron flow through the NADH dehydrogenase (Complex I), leading to ADP-stimulated oxygen consumption inhibition, transmembrane potential dissipation and cellular ATP level decrease, without increasing ROS production. Duroquinol, an electron donor at CoQ level, reversed the decrease of cell viability induced by HQ. Additionally, HQ selectively induced G₂/M-phase arrest. Taken together, our results suggest that the bioenergetic dysfunction provoked by HQ is implicated in its anticancer action.
Collapse
Affiliation(s)
- Félix A Urra
- Department of Organic and Physical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Sergio Livingstone 1007, Casilla 233, Santiago-1, Chile.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lin YW, Chiang BH. 4-acetylantroquinonol B isolated from Antrodia cinnamomea arrests proliferation of human hepatocellular carcinoma HepG2 cell by affecting p53, p21 and p27 levels. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:8625-8631. [PMID: 21739974 DOI: 10.1021/jf2011326] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The 4-acetylantroquinonol B isolated from the mycelium of Antrodia cinnamomea could inhibit proliferation of hepatocellular carcinoma cells HepG2 with IC(50) 0.1 μg/mL. When the HepG2 cells were treated with 4-acetylantroquinonol B for 72 h, the proportion of cells in the G1 phase of the cell cycle increased and that in the S phase decreased significantly, and the proportion of G2/M phase cells were not obviously changed. In addition, the 4-acetylantroquinonol B treatment resulted in the decreases of CDK2 and CDK4, and an increase of p27 in a dose-dependent manner. The protein levels of p53 and p21 proteins were also increased when the cells were treated with low dosage (0.1 μg/mL) of 4-acetylantroquinonol B. Higher dosages, however, decreased the expression of p53 and p21 proteins. Assay of RT-PCR indicated that, corresponding to the increases of p53 and p21 proteins at the dosage of 0.1 μg/mL, the mRNAs of p53 and p21 showed 1.66- and 1.61-fold upregulations, respectively. Corresponding to the decreases of CDK2 and CDK4 proteins, the mRNAs of CDK2 and CDK4 showed -1.02- and -1.13-fold downregulations, respectively. However, level of p27 mRNA showed -1.2-fold downregulation in spite of the increase in p27 protein. This observation, again, confirms the fact that the p27 gene rarely undergoes homozygous inactivation in cancer cells. Our finding suggested that the 4-acetylantroquinonol B inhibits proliferation of HepG2 cells via affecting p53, p21 and p27 proteins, and can be considered as a potential cancer drug.
Collapse
Affiliation(s)
- Yu-Wei Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
27
|
Papa L, Germain D. Estrogen receptor mediates a distinct mitochondrial unfolded protein response. J Cell Sci 2011; 124:1396-402. [PMID: 21486948 PMCID: PMC3078808 DOI: 10.1242/jcs.078220] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2011] [Indexed: 01/04/2023] Open
Abstract
Unfolded protein responses (UPRs) of the endoplasmic reticulum and mitochondrial matrix have been described. Here, we show that the accumulation of proteins in the inter-membrane space (IMS) of mitochondria in the breast cancer cell line MCF-7 activates a distinct UPR. Upon IMS stress, overproduction of reactive oxygen species (ROS) and phosphorylation of AKT triggers estrogen receptor (ER) activity, which further upregulates the transcription of the mitochondrial regulator NRF1 and the IMS protease OMI (officially known as HTRA2). Moreover, we demonstrate that the IMS stress-induced UPR culminates in increased proteasome activity. Given our previous report on a proteasome- and OMI-dependent checkpoint that limits the import of IMS proteins, the findings presented in this study suggest that this newly discovered UPR acts as a cytoprotective response to overcome IMS stress.
Collapse
Affiliation(s)
- Luena Papa
- Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, Box 1079, Mount Sinai School of Medicine, New York NY 10029, USA
| | - Doris Germain
- Tisch Cancer Institute, Department of Medicine, Division of Hematology/Oncology, Box 1079, Mount Sinai School of Medicine, New York NY 10029, USA
| |
Collapse
|
28
|
The anti-cancer agent nemorosone is a new potent protonophoric mitochondrial uncoupler. Mitochondrion 2010; 11:255-63. [PMID: 21044702 DOI: 10.1016/j.mito.2010.10.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 10/01/2010] [Accepted: 10/19/2010] [Indexed: 02/07/2023]
Abstract
Nemorosone, a natural-occurring polycyclic polyprenylated acylphloroglucinol, has received increasing attention due to its strong in vitro anti-cancer action. Here, we have demonstrated the toxic effect of nemorosone (1-25 μM) on HepG2 cells by means of the MTT assay, as well as early mitochondrial membrane potential dissipation and ATP depletion in this cancer cell line. In mitochondria isolated from rat liver, nemorosone (50-500 nM) displayed a protonophoric uncoupling activity, showing potency comparable to the classic protonophore, carbonyl cyanide m-chlorophenyl hydrazone (CCCP). Nemorosone enhanced the succinate-supported state 4 respiration rate, dissipated mitochondrial membrane potential, released Ca(2+) from Ca(2+)-loaded mitochondria, decreased Ca(2+) uptake and depleted ATP. The protonophoric property of nemorosone was attested by the induction of mitochondrial swelling in hyposmotic K(+)-acetate medium in the presence of valinomycin. In addition, uncoupling concentrations of nemorosone in the presence of Ca(2+) plus ruthenium red induced the mitochondrial permeability transition process. Therefore, nemorosone is a new potent protonophoric mitochondrial uncoupler and this property is potentially involved in its toxicity on cancer cells.
Collapse
|