1
|
Kim YM, Kim H, Park SC, Lee M, Jang MK. Targeted drug delivery of cancer cell-derived extracellular vesicles decorated with a VEGFR-binding peptide. Colloids Surf B Biointerfaces 2025; 252:114661. [PMID: 40203507 DOI: 10.1016/j.colsurfb.2025.114661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
Chemotherapy is commonly used to manage cancer lesions, but its use is limited or has a high risk of failure due to the side effects of the drugs. The use of a drug delivery system can minimize the side effects of drugs and maximize their anticancer effects. This study investigated the potential of tumor cell-derived small extracellular vesicles (sEV) as drug delivery vehicles for doxorubicin (Dox). In addition, the decoration of vascular endothelial growth factor receptor (VEGFR)-targeting peptide on the sEV provided an enhance specific cancer cell-targeting effect in vitro or homing capacity in vivo. The extrusion method was effective in loading Dox and displaying targeting peptide. The effective Dox release was resulted under acidic condition, an endosome pH. The growth of tumor masses on MCF-7 xenografted mice were significantly inhibited by this drug delivery system. We believe that our results will provide useful information for the development of chemotherapeutic drug delivery systems.
Collapse
Affiliation(s)
- Young-Min Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Hyeonseok Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Seong-Cheol Park
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| | - Mi-Kyeong Jang
- Department of Chemical Engineering, College of Engineering, Sunchon National University, 255 Jungangno, Suncheon, Jeonnam 57922, Republic of Korea.
| |
Collapse
|
2
|
Afriza D, Arma U, Faslah R, Suriyah WH. Anticancer Potential of Quercetin on Oral Squamous Cell Carcinoma: A Scoping Review and Molecular Docking. Eur J Dent 2025; 19:15-23. [PMID: 39348855 PMCID: PMC11750326 DOI: 10.1055/s-0044-1789016] [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: 10/02/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a type of cancer that has a low survival rate and high recurrence and metastasis rates. To date, there is still no effective treatment for OSCC. Various types of cancer, including OSCC, have reported quercetin to act as an anticancer agent, but there is no clear research data on how it may affect OSCC. To determine the anticancer potential of quercetin in OSCC, we conducted a scoping review, and to determine the interaction of quercetin with one of the proteins that plays a role in carcinogenesis, namely, BCL-2, we conducted molecular docking. The scoping review process was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews. The scoping review was searched by collecting articles related to the research topic in Google Scholar, PubMed, ScienceDirect, Cochrane, and EBSCOhost databases. All of the literature records found during the search were imported into the Mendeley software to remove duplication. Nine studies were generated after the titles and abstracts were reviewed according to the inclusion and exclusion criteria. After the full-text screening, no studies were excluded, leaving nine publications determined to be eligible for inclusion in the scoping review. Quercetin showed effects on inhibiting cancer invasion, migration, proliferation, and many protein expressions, as well as increasing cell apoptosis. Molecular docking was done for quercetin and BCl-2 protein. Doxorubicin was utilized as a comparison ligand. The in silico study was utilized using AutoDock Vina, AutoDock Tools 1.5.6, Biovia Discovery Studio 2021, and PyMol. Molecular docking indicated quercetin has a strong binding affinity with BCl-2 protein (ΔG -7.2 kcal/mol). Both scoping review and molecular docking revealed that quercetin is a promising candidate for anticancer agent.
Collapse
Affiliation(s)
- Dhona Afriza
- Department of Oral Biology, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | - Utmi Arma
- Department of Oral Medicine, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | - Raefany Faslah
- Department of Oral Medicine, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | | |
Collapse
|
3
|
Li H, Cheng S, Zhang Q, Zhou T, Zhang T, Liu S, Peng Y, Yu J, Xu J, Wang Q, Zhang J, Yao Y, Wang H. Dual-Multivalent Aptamer-Based Drug Delivery Platform for Targeted SRC Silencing to Enhance Doxorubicin Sensitivity in Endometrial Cancer. Int J Biol Sci 2024; 20:5812-5830. [PMID: 39664567 PMCID: PMC11628347 DOI: 10.7150/ijbs.101059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/19/2024] [Indexed: 12/13/2024] Open
Abstract
Endometrial cancer poses a significant threat to women's health. Doxorubicin is commonly used in chemotherapy for advanced and recurrent cases; however, low sensitivity frequently limits its effectiveness. In this study, we verified that SRC modulates the sensitivity of endometrial cancer to chemotherapy of doxorubicin and developed a targeted silencing drug delivery platform that employs rolling circle amplification and dual-multivalent aptamers to precisely deliver therapeutics directly to tumor cells. This platform enhanced endometrial cancer cell sensitivity to doxorubicin by modulating drug responsiveness at the genetic level. Our results suggest that this approach may improve cancer cell susceptibility to ferroptosis. The efficacy and safety of this platform were validated in both cellular and animal models. This study provides a new solution for realizing the precision treatment of endometrial cancer and lays a theoretical foundation for exploring the mechanism of endometrial cancer.
Collapse
Affiliation(s)
- Haojia Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Shuangshuang Cheng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Qi Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Ting Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Tangansu Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Shuangge Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Yingying Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Jia Yu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Jingwen Xu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Yuwei Yao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, China
- Clinical Research Center of Cancer Immunotherapy, Wuhan 430023, China
| |
Collapse
|
4
|
Szponar J, Gorska A, Ostrowska-Lesko M, Korga-Plewko A, Tchorz M, Ciechanski E, Dabrowska A, Poleszak E, Burdan F, Dudka J, Murias M, Mandziuk S. Assessment of the Impact of Carvedilol Administered Together with Dexrazoxan and Doxorubicin on Liver Structure and Function, Iron Metabolism, and Myocardial Redox System in Rats. Int J Mol Sci 2024; 25:2219. [PMID: 38396896 PMCID: PMC10889540 DOI: 10.3390/ijms25042219] [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: 01/06/2024] [Revised: 02/08/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Late cardiotoxicity is a formidable challenge in anthracycline-based anticancer treatments. Previous research hypothesized that co-administration of carvedilol (CVD) and dexrazoxane (DEX) might provide superior protection against doxorubicin (DOX)-induced cardiotoxicity compared to DEX alone. However, the anticipated benefits were not substantiated by the findings. This study focuses on investigating the impact of CVD on myocardial redox system parameters in rats treated with DOX + DEX, examining its influence on overall toxicity and iron metabolism. Additionally, considering the previously observed DOX-induced ascites, a seldom-discussed condition, the study explores the potential involvement of the liver in ascites development. Compounds were administered weekly for ten weeks, with a specific emphasis on comparing parameter changes between DOX + DEX + CVD and DOX + DEX groups. Evaluation included alterations in body weight, feed and water consumption, and analysis of NADPH2, NADP+, NADPH2/NADP+, lipid peroxidation, oxidized DNA, and mRNA for superoxide dismutase 2 and catalase expressions in cardiac muscle. The iron management panel included markers for iron, transferrin, and ferritin. Liver abnormalities were assessed through histological examinations, aspartate transaminase, alanine transaminase, and serum albumin level measurements. During weeks 11 and 21, reduced NADPH2 levels were observed in almost all examined groups. Co-administration of DEX and CVD negatively affected transferrin levels in DOX-treated rats but did not influence body weight changes. Ascites predominantly resulted from cardiac muscle dysfunction rather than liver-related effects. The study's findings, exploring the impact of DEX and CVD on DOX-induced cardiotoxicity, indicate a lack of scientific justification for advocating the combined use of these drugs at histological, biochemical, and molecular levels.
Collapse
Affiliation(s)
- Jaroslaw Szponar
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Agnieszka Gorska
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Marta Ostrowska-Lesko
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Medical University, 8b Jaczewski Street, 20-090 Lublin, Poland;
| | - Michal Tchorz
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Erwin Ciechanski
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
- Clinical Department of Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Anna Dabrowska
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodźko Street, 20-093 Lublin, Poland;
| | - Franciszek Burdan
- Human Anatomy Department, Medical University of Lublin, 4 Jaczewski Street, 20-090 Lublin, Poland;
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-608 Poznan, Poland;
| | - Slawomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland;
| |
Collapse
|
5
|
Beretta GL. Ferroptosis-induced Cardiotoxicity and Antitumor Drugs. Curr Med Chem 2024; 31:4935-4957. [PMID: 37469161 DOI: 10.2174/0929867331666230719124453] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
The induction of regulated cell death ferroptosis in tumors is emerging as an intriguing strategy for cancer treatment. Numerous antitumor drugs (e.g., doxorubicin, etoposide, tyrosine kinase inhibitors, trastuzumab, arsenic trioxide, 5-fluorouracil) induce ferroptosis. Although this mechanism of action is interesting for fighting tumors, the clinical use of drugs that induce ferroptosis is hampered by cardiotoxicity. Besides in cancer cells, ferroptosis induced by chemotherapeutics can occur in cardiomyocytes, and this feature represents an important drawback of antitumor therapy. This inconvenience has been tackled by developing less or no cardiotoxic antitumor drugs or by discovering cardioprotective agents (e.g., berberine, propofol, fisetin, salidroside, melatonin, epigallocatechin- 3gallate, resveratrol) to use in combination with conventional chemotherapeutics. This review briefly summarizes the molecular mechanisms of ferroptosis and describes the ferroptosis dependent mechanisms responsible for cardiac toxicity developed by cancer- suffering patients following the administration of some chemotherapeutics. Additionally, the pharmacological strategies very recently proposed for potentially preventing this inconvenience are considered.
Collapse
Affiliation(s)
- Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| |
Collapse
|
6
|
Jaiswara PK, Shukla SK. Chemotherapy-Mediated Neuronal Aberration. Pharmaceuticals (Basel) 2023; 16:1165. [PMID: 37631080 PMCID: PMC10459787 DOI: 10.3390/ph16081165] [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: 07/16/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Chemotherapy is a life-sustaining therapeutic option for cancer patients. Despite the advancement of several modern therapies, such as immunotherapy, gene therapy, etc., chemotherapy remains the first-line therapy for most cancer patients. Along with its anti-cancerous effect, chemotherapy exhibits several detrimental consequences that restrict its efficacy and long-term utilization. Moreover, it effectively hampers the quality of life of cancer patients. Cancer patients receiving chemotherapeutic drugs suffer from neurological dysfunction, referred to as chemobrain, that includes cognitive and memory dysfunction and deficits in learning, reasoning, and concentration ability. Chemotherapy exhibits neurotoxicity by damaging the DNA in neurons by interfering with the DNA repair system and antioxidant machinery. In addition, chemotherapy also provokes inflammation by inducing the release of various pro-inflammatory cytokines, including NF-kB, IL-1β, IL-6, and TNF-α. The chemotherapy-mediated inflammation contributes to chemobrain in cancer patients. These inflammatory cytokines modulate several growth signaling pathways and reactive oxygen species homeostasis leading to systemic inflammation in the body. This review is an effort to summarize the available information which discusses the role of chemotherapy-induced inflammation in chemobrain and how it impacts different aspects of therapeutic outcome and the overall quality of life of the patient. Further, this article also discusses the potential of herbal-based remedies to overcome chemotherapy-mediated neuronal toxicity as well as to improve the quality of life of cancer patients.
Collapse
Affiliation(s)
| | - Surendra Kumar Shukla
- Department of Oncology Science, University of Oklahoma Health Science Centre, Oklahoma City, OK 73104, USA;
| |
Collapse
|
7
|
Jeon J, Yoon B, Dey A, Song SH, Li Y, Joo H, Park JH. Self-immolative polymer-based immunogenic cell death inducer for regulation of redox homeostasis. Biomaterials 2023; 295:122064. [PMID: 36827894 DOI: 10.1016/j.biomaterials.2023.122064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
Doxorubicin (DOX), widely used as an anticancer drug, is considered an immunogenic cell death (ICD) inducer that enhances cancer immunotherapy. However, its extended application as an ICD inducer has been limited owing to poor antigenicity and inefficient adjuvanticity. To enhance the immunogenicity of DOX, we prepare a reactive oxygen species (ROS)-responsive self-immolative polymer (R-SIP) that can efficiently destroy redox homeostasis via self-immolation-mediated glutathione depletion in cancer cells. Owing to its amphiphilic nature, R-SIP self-assemble into nano-sized particles under aqueous conditions, and DOX is efficiently encapsulated inside the nanoparticles by a simple dialysis method. Interestingly, when treated with 4T1 cancer cells, DOX-encapsulated R-SIP (DR-SIP) induces the phosphorylation of eukaryotic translation initiation factor 2α and overexpression of ecto-calreticulin, resulting in endoplasmic reticulum-associated ICD. In addition, DR-SIP contributes to the maturation of dendritic cells by promoting the release of damage-associated molecular patterns (DAMPs) from cancer cells. When intravenously administered to tumor-bearing mice, DR-SIP remarkably inhibits tumor growth compared with DOX alone. Overall, DR-SIP may have the potential to elicit an immune response as an ICD inducer.
Collapse
Affiliation(s)
- Jueun Jeon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Been Yoon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Anup Dey
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seok Ho Song
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yuce Li
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyeyeon Joo
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
8
|
Understanding the Protective Role of Exosomes in Doxorubicin-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2852251. [PMID: 36132225 PMCID: PMC9484956 DOI: 10.1155/2022/2852251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 12/06/2022]
Abstract
Doxorubicin (DOX) is a class of effective chemotherapeutic agents widely used in clinical practice. However, its use has been limited by cardiotoxicity. The mechanism of DOX-induced cardiotoxicity (DIC) is complex, involving oxidative stress, Ca2+ overload, inflammation, pyroptosis, ferroptosis, apoptosis, senescence, etc. Exosomes (EXOs), as extracellular vesicles (EVs), play an important role in the material exchange and signal transmission between cells by carrying components such as proteins and RNAs. More recently, there has been a growing number of publications focusing on the protective effect of EXOs on DIC. Here, this review summarized the main mechanisms of DIC, discussed the mechanism of EXOs in the treatment of DIC, and further explored the value of EXOs as diagnostic biomarkers and therapeutic strategies for DIC.
Collapse
|
9
|
Zhang G, Yuan C, Su X, Zhang J, Gokulnath P, Vulugundam G, Li G, Yang X, An N, Liu C, Sun W, Chen H, Wu M, Sun S, Xing Y. Relevance of Ferroptosis to Cardiotoxicity Caused by Anthracyclines: Mechanisms to Target Treatments. Front Cardiovasc Med 2022; 9:896792. [PMID: 35770215 PMCID: PMC9234116 DOI: 10.3389/fcvm.2022.896792] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 12/06/2022] Open
Abstract
Anthracyclines (ANTs) are a class of anticancer drugs widely used in oncology. However, the clinical application of ANTs is limited by their cardiotoxicity. The mechanisms underlying ANTs-induced cardiotoxicity (AIC) are complicated and involve oxidative stress, inflammation, topoisomerase 2β inhibition, pyroptosis, immunometabolism, autophagy, apoptosis, ferroptosis, etc. Ferroptosis is a new form of regulated cell death (RCD) proposed in 2012, characterized by iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation. An increasing number of studies have found that ferroptosis plays a vital role in the development of AIC. Therefore, we aimed to elaborate on ferroptosis in AIC, especially by doxorubicin (DOX). We first summarize the mechanisms of ferroptosis in terms of oxidation and anti-oxidation systems. Then, we discuss the mechanisms related to ferroptosis caused by DOX, particularly from the perspective of iron metabolism of cardiomyocytes. We also present our research on the prevention and treatment of AIC based on ferroptosis. Finally, we enumerate our views on the development of drugs targeting ferroptosis in this emerging field.
Collapse
Affiliation(s)
- Guoxia Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chao Yuan
- Dezhou Second People’s Hospital, Dezhou, China
| | - Xin Su
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianzhen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Gururaja Vulugundam
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Naples, Italy
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xinyu Yang
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Can Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wanli Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hengwen Chen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shipeng Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Shipeng Sun,
| | - Yanwei Xing
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Yanwei Xing,
| |
Collapse
|
10
|
Yousefian M, Hosseinzadeh H, Hayes AW, Hadizadeh F, Karimi G. The Protective Effect of Natural Compounds on Doxorubicin-Induced Cardiotoxicity via Nicotinamide Adenine Dinucleotide Phosphate Oxidase Inhibition. J Pharm Pharmacol 2021; 74:351-359. [PMID: 34562089 DOI: 10.1093/jpp/rgab109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 07/12/2021] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Doxorubicin (DOX) is widely prescribed for the treatment of several human cancers. Unfortunately, cumulative doses of DOX are the main cause of myocardial dysfunction. Although preclinical and pharmaceutical studies were performed to investigate the potential of natural compounds in minimizing DOX toxicity, a comprehensive review of them is not available. This review can help the researchers for an effective search strategy. KEY FINDINGS Oxidative stress and p53 play an important role in DOX-associated cardiotoxicity. DOX activates nicotinamide adenine dinucleotide phosphate NADPH oxidase (NOX) in the heart, resulting in excessive reactive oxygen species that can induce cardiomyocyte apoptosis through phosphorylation of p53, DNA damage and/or mitogen-activated protein kinases-mediated cardiomyocyte apoptosis. Although a few chemical drugs with high efficacy are administered along with DOX to prevent or more likely to reduce cardiovascular toxicity, their use is often limited by additional side effects. Recently, attention has been drawn to natural compounds that prevent DOX cardiotoxicity. This review focuses on some of the natural bioactive compounds with potential therapeutic efficacy against DOX-induced cardiotoxicity (DIC). SUMMARY Some natural compounds, especially flavonols, flavonoids and proanthocyanidins, have the most protective effects against DIC by forming stable radicals and preventing the assembly of the NOX subunits.
Collapse
Affiliation(s)
- Mozhdeh Yousefian
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental/Occupational Risk Analysis & Management, University of South Florida, College of Public Health, Tampa, FLUSA.,Michigan State University, East Lansing, MI, USA
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Systems Biology Approaches to Enzyme Kinetics. Methods Mol Biol 2021. [PMID: 34272703 DOI: 10.1007/978-1-0716-1554-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Intracellular drug metabolism involves transport, bioactivation, conjugation, and other biochemical steps. The dynamics of these steps are each dependent on a number of other cellular factors that can ultimately lead to unexpected behavior. In this review, we discuss the confounding processes and coupled reactions within bioactivation networks that require a systems-level perspective in order to fully understand the time-varying behavior. When converting known in vitro characteristics of drug-enzyme interactions into descriptions of cellular systems, features such as substrate availability, cell-to-cell variability, and intracellular redox state, deserve special focus. Two examples are provided. First, a model of hydrogen peroxide clearance during chemotherapy treatment serves as a basis to discuss an example of sensitivity analysis. Second, an example of doxorubicin bioactivation is used for discussing points of consideration when constructing and analyzing network models of drug metabolism.
Collapse
|
12
|
Elblehi SS, El-Sayed YS, Soliman MM, Shukry M. Date Palm Pollen Extract Avert Doxorubicin-Induced Cardiomyopathy Fibrosis and Associated Oxidative/Nitrosative Stress, Inflammatory Cascade, and Apoptosis-Targeting Bax/Bcl-2 and Caspase-3 Signaling Pathways. Animals (Basel) 2021; 11:ani11030886. [PMID: 33804672 PMCID: PMC8003775 DOI: 10.3390/ani11030886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The use of date palm pollen ethanolic extract (DPPE) is a conventional approach in improving the side-effects induced by Doxorubicin (DOX).DPPE mitigated DOX-induced body and heart weight changes and ameliorated DOX-induced elevated cardiac injury markers. In addition, serum cardiac troponin I concentrations (cTnI), troponin T (cTnT), and N-terminal NBP and cytosolic (Ca+2) were amplified by alleviating the inflammatory and oxidative injury markers and decreasing histopathological lesions severity. DPPE decreased DOX-induced heart injuries by mitigating inflammation, fibrosis, and apoptosis through its antioxidant effect. To reduce DOX-induced oxidative stress injuries and other detrimental effects, a combined treatment of DPPE is advocated. Abstract Doxorubicin (DOX) has a potent antineoplastic efficacy and is considered a cornerstone of chemotherapy. However, it causes several dose-dependent cardiotoxic results, which has substantially restricted its clinical application. This study was intended to explore the potential ameliorative effect of date palm pollen ethanolic extract (DPPE) against DOX-induced cardiotoxicity and the mechanisms underlying it. Forty male Wistar albino rats were equally allocated into Control (CTR), DPPE (500 mg/kg bw for 4 weeks), DOX (2.5 mg/kg bw, intraperitoneally six times over 2 weeks), and DPPE + DOX-treated groups. Pre-coadministration of DPPE with DOX partially ameliorated DOX-induced cardiotoxicity as DPPE improved DOX-induced body and heart weight changes and mitigated the elevated cardiac injury markers activities of serum aminotransferases, lactate dehydrogenase, creatine kinase, and creatine kinase-cardiac type isoenzyme. Additionally, the concentration of serum cardiac troponin I (cTnI), troponin T (cTnT), N-terminal pro-brain natriuretic peptide (NT-pro BNP), and cytosolic calcium (Ca+2) were amplified. DPPE also alleviated nitrosative status (nitric oxide) in DOX-treated animals, lipid peroxidation and antioxidant molecules as glutathione content, and glutathione peroxidase, catalase, and superoxide dismutase activities and inflammatory markers levels; NF-κB p65, TNF-α, IL-1β, and IL-6. As well, it ameliorated the severity of histopathological lesions, histomorphometric alteration and improved the immune-staining of the pro-fibrotic (TGF-β1), pro-apoptotic (caspase-3 and Bax), and anti-apoptotic (Bcl-2) proteins in cardiac tissues. Collectively, pre-coadministration of DPPE partially mitigated DOX-induced cardiac injuries via its antioxidant, anti-inflammatory, anti-fibrotic, and anti-apoptotic potential.
Collapse
Affiliation(s)
- Samar S. Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Edfina 22758, Egypt
- Correspondence: (S.S.E.); (M.S.)
| | - Yasser S. El-Sayed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
- Correspondence: (S.S.E.); (M.S.)
| |
Collapse
|
13
|
Salsabila IA, Nugraheni N, Ahlina FN, Haryanti S, Meiyanto E. Synergistic Cotreatment Potential of Soursop ( Annona muricata L.) Leaves Extract with Doxorubicin on 4T1 Cells with Antisenescence and Anti-reactive-oxygen-species Properties. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:57-67. [PMID: 34567146 PMCID: PMC8457719 DOI: 10.22037/ijpr.2020.112485.13788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Annona muricata L. extract (AME) exhibits cytotoxic activities on various types of cancer cells. This study aims to unveil the anticancer activity of AME as a cotreatment agent with doxorubicin (dox) on 4T1 cells and AME's relation to senescence. AME was obtained by maceration using 96% ethanol. AME was then subjected to qualitative analysis using TLC compared to quercetin (hRf = 75). Spectrophotometry analysis of AME resulted in a total flavonoid content of 2.3% ± 0.05%. Cytotoxic evaluation using the MTT assay revealed that AME showed an IC50 value of 63 µg/mL, while its combination (25 µg/mL) with dox (10 nM) decreased the viability of 4T1 cells to 58 % (CI = 0.15). Flowcytometry using propidium iodide staining confirmed that AME (13 and 25 µg/mL) caused cell cycle arrest in the G1 phase as a single treatment and G2/M arrest in combination with dox. However, by using the dichloro dihydrofluorescein diacetate staining assay, it turned out that AME at concentrations of 13 and 25 µg/mL decreased intracellular reactive oxygen species (ROS) levels both as a single treatment and in combination with dox. Senescence-associated β - galactosidase assay showed that AME decreased dox-induced senescence. AME alone and in combination with dox (cotreatment) showed cytotoxic effect synergistically on 4T1 cells, but this was not caused by an increase in intracellular ROS levels as well as senescence induction. Therefore, AME showed its potential to be a cotreatment agent with antioxidant property on triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Irfani Aura Salsabila
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Nadzifa Nugraheni
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Faradiba Nur Ahlina
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Sari Haryanti
- Medicinal Plant and Traditional Medicinal Research and Development Centre, Ministry of Health, Republic of Indonesia, Tawangmangu, Central Java, Indonesia.
| | - Edy Meiyanto
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
- Laboratory of Macromolecular Engineering Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Yogyakarta, Indonesia.
| |
Collapse
|
14
|
Yang L, Zhang C, Chen J, Zhang S, Pan G, Xin Y, Lin L, You Z. Shenmai injection suppresses multidrug resistance in MCF-7/ADR cells through the MAPK/NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2020; 58:276-285. [PMID: 32251615 PMCID: PMC7170370 DOI: 10.1080/13880209.2020.1742167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Context: Shenmai Injection (SMI) is usually used to treat atherosclerotic coronary heart disease and viral myocarditis in China. However, the effect of SMI on multidrug resistance has not been reported.Objective: To investigate the reversal effect of SMI in adriamycin (ADR) resistant breast cancer cell line (MCF-7/ADR) and explore the related molecular mechanisms.Materials and methods: The effect of SMI (0.25, 0.5, 1 mg/mL) to reverse chemoresistance in MCF-7/ADR cells was elucidated by MTT, HPLC-FLD, DAPI staining, flow cytometric analysis, western blotting. At the same time, in vivo test was conducted to probe into the effect of SMI on reversing ADR resistance, and verapamil (10 μM) was used as a positive control.Results: The results showed that the toxicity of ADR to MCF-7/ADR cells was strengthened significantly after treated with SMI (0.25, 0.5, 1 mg/mL), the IC50 of ADR was decreased 54.4-fold. The intracellular concentrations of ADR were increased 2.2-fold (p < 0.05) and ADR accumulation was enhanced in the nuclei (p < 0.05). SMI could strongly enhance the ADR-induced apoptosis and increase intracellular rhodamine 123 accumulation in MCF-7/ADR cells. Additionally, a combination of ADR and SMI (5 mg/kg) could dramatically reduce the weight and volume of tumour (p < 0.05). Furthermore, the results revealed that SMI might reverse MDR via inhibiting ADR-induced activation of the mitogen-activated protein kinase/nuclear factor (NF)-κB pathway to down-regulated the expression of P-glycoprotein (P-gp).Discussion and conclusions: SMI could potentially be used to treat ADR-resistance. This suggests possibilities for future clinical research.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/metabolism
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Down-Regulation/drug effects
- Doxorubicin/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Combinations
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Humans
- MAP Kinase Signaling System/drug effects
- MCF-7 Cells
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- Rhodamine 123/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lin Yang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Chengda Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Jiaoting Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Sheng Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Guixuan Pan
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yanfei Xin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
- Yanfei Xin
| | - Lin Lin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Lin Lin Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou310013, China
| | - Zhenqiang You
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- CONTACT Zhenqiang You
| |
Collapse
|
15
|
Najafi M, Mortezaee K, Rahimifard M, Farhood B, Haghi-Aminjan H. The role of curcumin/curcuminoids during gastric cancer chemotherapy: A systematic review of non-clinical study. Life Sci 2020; 257:118051. [DOI: 10.1016/j.lfs.2020.118051] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
|
16
|
Protective Effect of Methylxanthine Fractions Isolated from Bancha Tea Leaves against Doxorubicin-Induced Cardio- and Nephrotoxicities in Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4018412. [PMID: 32851069 PMCID: PMC7439203 DOI: 10.1155/2020/4018412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/18/2020] [Accepted: 07/21/2020] [Indexed: 11/18/2022]
Abstract
Doxorubicin is an anthracycline antibiotic that is used for the treatment of various types of cancer. However, its clinical usage is limited due to its potential life-threatening adverse effects, such as cardio- and nephrotoxicities. Nonetheless, simultaneous administration of doxorubicin and antioxidants, such as those found in green tea leaves, could reduce cardiac and renal tissue damage caused by oxidative stress. The methylxanthine fraction isolated from Bancha tea leaves were tested in vitro for its antioxidant activity and in vivo for its organoprotective properties against doxorubicin-induced cardio- and nephrotoxicities in a rat model. The in vivo study was conducted on male Wistar rats divided into 6 groups. Methylxanthines were administered at high (5 mg/kg body weight) and low (1 mg/kg body weight) doses, while doxorubicin was administered at a cumulative dose of 20 mg/kg body weight. Serum creatinine, uric acid, and urea concentrations, as well as serum enzyme levels (creatinine kinase (CK), creatinine kinase MB fraction (CK-MB), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH)) and electrolytes (Na+, K+, and Cl−), were analysed. In addition, histological analysis was performed to assess cardiac and renal tissue damage. The concomitant administration of Bancha methylxanthines and doxorubicin showed a dose-dependent reduction in the serum biochemical parameters, indicating a decrease in the cardiac and renal tissue damage caused by the antibiotic. Histological analysis showed that pretreatment with methylxanthines at the dose of 5 mg/kg resulted in an almost normal myocardial structure and a significant decrease in the morphological kidney changes caused by doxorubicin exposure compared with the group that received doxorubicin alone. The putative mechanism is most likely related to a reduction in the oxidative stress caused by doxorubicin.
Collapse
|
17
|
Clerodendrum volubile Ethanol Leaf Extract: A Potential Antidote to Doxorubicin-Induced Cardiotoxicity in Rats. J Toxicol 2020; 2020:8859716. [PMID: 32714390 PMCID: PMC7355376 DOI: 10.1155/2020/8859716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/11/2020] [Indexed: 01/09/2023] Open
Abstract
Doxorubicin is widely applied in hematological and solid tumor treatment but limited by its off-target cardiotoxicity. Thus, cardioprotective potential and mechanism(s) of CVE in DOX-induced cardiotoxicity were investigated using cardiac and oxidative stress markers and histopathological endpoints. 50–400 mg/kg/day CVE in 5% DMSO in distilled water were investigated in Wistar rats intraperitoneally injected with 2.5 mg/kg DOX on alternate days for 14 days, using serum troponin I and LDH, complete lipid profile, cardiac tissue oxidative stress marker assays, and histopathological examination of DOX-treated cardiac tissue. Preliminary qualitative and quantitative assays of CVE's secondary metabolites were also conducted. Phytochemical analyses revealed the presence of flavonoids (34.79 ± 0.37 mg/100 mg dry extract), alkaloids (36.73 ± 0.27 mg/100 mg dry extract), reducing sugars (07.78 ± 0.09 mg/100 mg dry extract), and cardiac glycosides (24.55 ± 0.12 mg/100 mg dry extract). 50–400 mg/kg/day CVE significantly attenuated increases in the serum LDH and troponin I levels. Similarly, the CVE dose unrelatedly decreased serum TG and VLDL-c levels without significant alterations in the serum TC, HDL-c, and LDL-c levels. Also, CVE profoundly attenuated alterations in the cardiac tissue oxidative stress markers' activities while improving DOX-associated cardiac histological lesions that were possibly mediated via free radical scavenging and/or antioxidant mechanisms. Overall, CVE may play a significant therapeutic role in the management of DOX-induced cardiotoxicity in humans.
Collapse
|
18
|
Held JM. Redox Systems Biology: Harnessing the Sentinels of the Cysteine Redoxome. Antioxid Redox Signal 2020; 32:659-676. [PMID: 31368359 PMCID: PMC7047077 DOI: 10.1089/ars.2019.7725] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022]
Abstract
Significance: Cellular redox processes are highly interconnected, yet not in equilibrium, and governed by a wide range of biochemical parameters. Technological advances continue refining how specific redox processes are regulated, but broad understanding of the dynamic interconnectivity between cellular redox modules remains limited. Systems biology investigates multiple components in complex environments and can provide integrative insights into the multifaceted cellular redox state. This review describes the state of the art in redox systems biology as well as provides an updated perspective and practical guide for harnessing thousands of cysteine sensors in the redoxome for multiparameter characterization of cellular redox networks. Recent Advances: Redox systems biology has been applied to genome-scale models and large public datasets, challenged common conceptions, and provided new insights that complement reductionist approaches. Advances in public knowledge and user-friendly tools for proteome-wide annotation of cysteine sentinels can now leverage cysteine redox proteomics datasets to provide spatial, functional, and protein structural information. Critical Issues: Careful consideration of available analytical approaches is needed to broadly characterize the systems-level properties of redox signaling networks and be experimentally feasible. The cysteine redoxome is an informative focal point since it integrates many aspects of redox biology. The mechanisms and redox modules governing cysteine redox regulation, cysteine oxidation assays, proteome-wide annotation of the biophysical and biochemical properties of individual cysteines, and their clinical application are discussed. Future Directions: Investigating the cysteine redoxome at a systems level will uncover new insights into the mechanisms of selectivity and context dependence of redox signaling networks.
Collapse
Affiliation(s)
- Jason M. Held
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| |
Collapse
|
19
|
Boussada M, Ali RB, Chahbi A, Abdelkarim M, Fradj MKB, Dziri C, Bokri K, Akacha AB, El May MV. A new Thiocyanoacetamide protects rat sperm cells from Doxorubicin-triggered cytotoxicity whereas Selenium shows low efficacy: In vitro approach. Toxicol In Vitro 2019; 61:104587. [PMID: 31271807 DOI: 10.1016/j.tiv.2019.06.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/29/2019] [Accepted: 06/29/2019] [Indexed: 11/26/2022]
Abstract
Doxorubicin (DOX) exhibits a wide-ranging spectrum of antitumor activities which maintain its clinical use despite its devastating impact on highly proliferating cells. The present work was designed to develop a new approach which aims to protect male germ cells from DOX cytotoxicity. Thus, an assessment of the protective potential of a new thioamide analog (thiocyanoacetamide; TA) compared to selenium (Se) was performed in rat sperms exposed to DOX in vitro. Oxygen consumption rate (OCR) was measured after exposure to three different doses (0.5, 1, 1.5 and 2 μM) of DOX, Se or TA, and the suitable concentrations were selected for further studies afterwards. Motility, OCR in a time-dependent manner, glucose extracellular concentration and lipid peroxidation (LPO) were measured. Fatty acid (FA) content was assessed by gas chromatography (GC-FID). Cell death, superoxide anion (O2-), mitochondrial membrane potential (MMP), and DNA damage were evaluated by flow cytometry. TA association with DOX increased OCR and glucose uptake, improved cell survival and decreased DNA damage. The co-administration of DOX with Se increased OCR, significantly prevented O2- overproduction, and decreased LPO. Collected data brought new insights regarding this transformed TA, which showed better efficiency than Se in reducing DOX cytotoxic stress in sperms.
Collapse
Affiliation(s)
- Marwa Boussada
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Ridha Ben Ali
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia; Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Ahlem Chahbi
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Mohamed Abdelkarim
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Mohamed Kacem Ben Fradj
- UR05/08-08, LR99/ES/11, Department of Biochemistry, Rabta Hospital, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Chadli Dziri
- Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Khouloud Bokri
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Azaiez Ben Akacha
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Michèle Véronique El May
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| |
Collapse
|
20
|
Grebinyk A, Prylutska S, Chepurna O, Grebinyk S, Prylutskyy Y, Ritter U, Ohulchanskyy TY, Matyshevska O, Dandekar T, Frohme M. Synergy of Chemo- and Photodynamic Therapies with C 60 Fullerene-Doxorubicin Nanocomplex. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1540. [PMID: 31671590 PMCID: PMC6915635 DOI: 10.3390/nano9111540] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 10/27/2019] [Indexed: 12/17/2022]
Abstract
A nanosized drug complex was explored to improve the efficiency of cancer chemotherapy, complementing it with nanodelivery and photodynamic therapy. For this, nanomolar amounts of a non-covalent nanocomplex of Doxorubicin (Dox) with carbon nanoparticle C60 fullerene (C60) were applied in 1:1 and 2:1 molar ratio, exploiting C60 both as a drug-carrier and as a photosensitizer. The fluorescence microscopy analysis of human leukemic CCRF-CEM cells, in vitro cancer model, treated with nanocomplexes showed Dox's nuclear and C60's extranuclear localization. It gave an opportunity to realize a double hit strategy against cancer cells based on Dox's antiproliferative activity and C60's photoinduced pro-oxidant activity. When cells were treated with 2:1 C60-Dox and irradiated at 405 nm the high cytotoxicity of photo-irradiated C60-Dox enabled a nanomolar concentration of Dox and C60 to efficiently kill cancer cells in vitro. The high pro-oxidant and pro-apoptotic efficiency decreased IC50 16, 9 and 7 × 103-fold, if compared with the action of Dox, non-irradiated nanocomplex, and C60's photodynamic effect, correspondingly. Hereafter, a strong synergy of therapy arising from the combination of C60-mediated Dox delivery and C60 photoexcitation was revealed. Our data indicate that a combination of chemo- and photodynamic therapies with C60-Dox nanoformulation provides a promising synergetic approach for cancer treatment.
Collapse
Affiliation(s)
- Anna Grebinyk
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| | - Svitlana Prylutska
- Taras Shevchenko National University of Kyiv, Volodymyrska 64, 01601 Kyiv, Ukraine.
| | - Oksana Chepurna
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Sergii Grebinyk
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
| | - Yuriy Prylutskyy
- Taras Shevchenko National University of Kyiv, Volodymyrska 64, 01601 Kyiv, Ukraine.
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, University of Technology Ilmenau, Weimarer Straße 25 (Curiebau), 98693 Ilmenau, Germany.
| | - Tymish Y Ohulchanskyy
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Olga Matyshevska
- Palladin Institute of Biochemistry, NAS of Ukraine, Leontovicha Str. 9, 01030 Kyiv, Ukraine.
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| | - Marcus Frohme
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
| |
Collapse
|
21
|
Lavogina D, Samuel K, Lavrits A, Meltsov A, Sõritsa D, Kadastik Ü, Peters M, Rinken A, Salumets A. Chemosensitivity and chemoresistance in endometriosis – differences for ectopic versus eutopic cells. Reprod Biomed Online 2019; 39:556-568. [DOI: 10.1016/j.rbmo.2019.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 01/19/2023]
|
22
|
Koleini N, Nickel BE, Edel AL, Fandrich RR, Ravandi A, Kardami E. Oxidized phospholipids in Doxorubicin-induced cardiotoxicity. Chem Biol Interact 2019; 303:35-39. [DOI: 10.1016/j.cbi.2019.01.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 01/31/2023]
|
23
|
Chen X, Mims J, Huang X, Singh N, Motea E, Planchon SM, Beg M, Tsang AW, Porosnicu M, Kemp ML, Boothman DA, Furdui CM. Modulators of Redox Metabolism in Head and Neck Cancer. Antioxid Redox Signal 2018; 29:1660-1690. [PMID: 29113454 PMCID: PMC6207163 DOI: 10.1089/ars.2017.7423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Head and neck squamous cell cancer (HNSCC) is a complex disease characterized by high genetic and metabolic heterogeneity. Radiation therapy (RT) alone or combined with systemic chemotherapy is widely used for treatment of HNSCC as definitive treatment or as adjuvant treatment after surgery. Antibodies against epidermal growth factor receptor are used in definitive or palliative treatment. Recent Advances: Emerging targeted therapies against other proteins of interest as well as programmed cell death protein 1 and programmed death-ligand 1 immunotherapies are being explored in clinical trials. CRITICAL ISSUES The disease heterogeneity, invasiveness, and resistance to standard of care RT or chemoradiation therapy continue to constitute significant roadblocks for treatment and patients' quality of life (QOL) despite improvements in treatment modality and the emergence of new therapies over the past two decades. FUTURE DIRECTIONS As reviewed here, alterations in redox metabolism occur at all stages of HNSCC management, providing opportunities for improved prevention, early detection, response to therapies, and QOL. Bioinformatics and computational systems biology approaches are key to integrate redox effects with multiomics data from cells and clinical specimens and to identify redox modifiers or modifiable target proteins to achieve improved clinical outcomes. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jade Mims
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiumei Huang
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Naveen Singh
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Edward Motea
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | | | - Muhammad Beg
- Department of Internal Medicine, Division of Hematology-Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Melissa L. Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - David A. Boothman
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
24
|
Lewis JE, Costantini F, Mims J, Chen X, Furdui CM, Boothman DA, Kemp ML. Genome-Scale Modeling of NADPH-Driven β-Lapachone Sensitization in Head and Neck Squamous Cell Carcinoma. Antioxid Redox Signal 2018; 29:937-952. [PMID: 28762750 PMCID: PMC6104251 DOI: 10.1089/ars.2017.7048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS The purpose of this study was to investigate differential nicotinamide adenine dinucleotide phosphate, reduced (NADPH) production between radiation-sensitive and -resistant head and neck squamous cell carcinoma (HNSCC) cell lines and whether these differences are predictive of sensitivity to the chemotherapeutic β-lapachone. RESULTS We have developed a novel human genome-scale metabolic modeling platform that combines transcriptomic, kinetic, thermodynamic, and metabolite concentration data. Upon incorporation of this information into cell line-specific models, we observed that the radiation-resistant HNSCC model redistributed flux through several major NADPH-producing reactions. Upon RNA interference of canonical NADPH-producing genes, the metabolic network can further reroute flux through alternate NADPH biosynthesis pathways in a cell line-specific manner. Model predictions of perturbations in cellular NADPH production after gene knockdown match well with experimentally verified effects of β-lapachone treatment on NADPH/NADP+ ratio and cell viability. This computational approach accurately predicts HNSCC-specific oxidoreductase genes that differentially affect cell viability between radiation-responsive and radiation-resistant cancer cells upon β-lapachone treatment. INNOVATION Quantitative genome-scale metabolic models that incorporate multiple levels of biological data are applied to provide accurate predictions of responses to a NADPH-dependent redox cycling chemotherapeutic drug under a variety of perturbations. CONCLUSION Our modeling approach suggests differences in metabolism and β-lapachone redox cycling that underlie phenotypic differences in radiation-sensitive and -resistant cancer cells. This approach can be extended to investigate the synergistic action of NAD(P)H: quinone oxidoreductase 1 bioactivatable drugs and radiation therapy. Antioxid. Redox Signal. 29, 937-952.
Collapse
Affiliation(s)
- Joshua E Lewis
- 1 The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| | - Francesco Costantini
- 2 School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Jade Mims
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Xiaofei Chen
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Cristina M Furdui
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - David A Boothman
- 4 Department of Pharmacology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Melissa L Kemp
- 1 The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| |
Collapse
|
25
|
Wu S, Chou H, Yuh C, Mekuria SL, Kao Y, Tsai H. Radiation-Sensitive Dendrimer-Based Drug Delivery System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700339. [PMID: 29610720 PMCID: PMC5827102 DOI: 10.1002/advs.201700339] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/10/2017] [Indexed: 05/13/2023]
Abstract
Combination of chemotherapy and radiotherapy is used to enhance local drug delivery while reducing off-target tissue effects. Anticancer drug doxorubicin (DOX) is loaded into l-cysteine modified G4.5 dendrimer (GC/DOX) and released at different pH values in the presence and absence of γ-radiation. Presence of γ-radiation significantly improves DOX release from the GC/DOX under acidic pH conditions, suggesting that GC dendrimer is a radiation-sensitive drug delivery system. GC/DOX is further evaluated by determining cytotoxicity in uterine cervical carcinoma HeLa cells. GC/DOX shows high affinity for cancer cells and effective drug release following an external stimulus (radiation exposure), whereas an in vivo zebrafish study confirms that l-cysteine acts as a radiosensitizer. GC/DOX treatment combined with radiotherapy synergistically and successfully inhibits cancer cell growth.
Collapse
Affiliation(s)
- Szu‐Yuan Wu
- Department of Radiation OncologyWan Fang HospitalTaipei Medical University116TaipeiTaiwan
- Department of Internal MedicineSchool of MedicineCollege of MedicineTaipei Medical University110TaipeiTaiwan
| | - Hsiao‐Ying Chou
- Graduate Institute of Applied Science and TechnologyNational Taiwan University of Science and Technology106TaipeiTaiwan
| | - Chiou‐Hwa Yuh
- Institute of Molecular and Genomic MedicineNational Health Research Institutes350ZhunanMiaoliTaiwan
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua University300HsinchuTaiwan
- Department of Biological Science and TechnologyNational Chiao Tung University300HsinchuTaiwan
| | - Shewaye Lakew Mekuria
- Graduate Institute of Applied Science and TechnologyNational Taiwan University of Science and Technology106TaipeiTaiwan
| | - Yu‐Chih Kao
- Graduate Institute of Applied Science and TechnologyNational Taiwan University of Science and Technology106TaipeiTaiwan
| | - Hsieh‐Chih Tsai
- Graduate Institute of Applied Science and TechnologyNational Taiwan University of Science and Technology106TaipeiTaiwan
| |
Collapse
|
26
|
Alves Gomes FT, Boleti APDA, Leandro LM, Squinello D, Aranha ESP, Vasconcelos MC, Cos P, Veiga-Junior VF, Lima ES. Biological Activities and Cytotoxicity of Eperua oleifera Ducke Oil-resin. Pharmacogn Mag 2017; 13:542-552. [PMID: 29200711 PMCID: PMC5701389 DOI: 10.4103/pm.pm_552_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/01/2017] [Indexed: 01/09/2023] Open
Abstract
Background The oil-resin of Eperua oleifera Ducke has been used in popular medicine similarly to the copaiba oil (Copaifera spp.). Objective This study aimed to investigate the effects of the acid fraction of E. oleifera oil-resin (AFEOR) on cell proliferation, collagen production in human fibroblasts, inhibition of metalloproteinases, and cytotoxicity against tumor cell lines. Materials and Methods Acid fraction of E. oleifera was fractionated in the ion exchange column chromatography. Cytotoxicity and genotoxicity were evaluated by Alamar Blue® and Cometa assay. The inhibition of metalloproteinases was performed by zymography and Western blotting. Results The predominant acidic diterpenes in the AFEOR were copalic and hardwickiic acids. AFEOR caused morphology alteration and decrease of proliferation at concentrations higher than 5 μg/mL. It also caused significant collagen proliferation in fibroblasts. It showed cytotoxicity against tumoral and nontumoral cell lines, with IC50 values ranging from 13 to 50 μg/mL, and a hemolytic activity with an IC50 value of 38.29 μg/mL. AFEOR inhibited collagenase activity, with an IC50 value of 46.64 μg/mL, and matrix metalloproteinase-2 (MMP)-2 and MMP-9 in HaCaT cells or MMP-1 expression in MRC-5 cells. AFEOR induced genotoxicity in MRC-5 cells with a DNA damage index between 40% and 60% when compared to the negative controls (0%-20%). Conclusion For the first time, biological activities from oil-resin E. oleifera demonstrated ratifying somehow its popular use. SUMMARY Analysis of crude oil-resin and fractionation of diterpenic fraction was performance using selective ion-exchange column chromatographyCytotoxicity analysis and morphology were performed with different cell linesCollagen production in human fibroblasts, inhibition of metalloproteinases were demonstrated by zymography and Western blotting. Abbreviations used: AFEOR: Eperua oleifera oil-resin.
Collapse
Affiliation(s)
- Fernanda Torlania Alves Gomes
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Ana Paula de Araújo Boleti
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Lidiam M Leandro
- Chemistry Department, Exact Sciences Institute, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Diego Squinello
- Chemistry Department, Exact Sciences Institute, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Ellen S P Aranha
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Marne C Vasconcelos
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Paul Cos
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, B2610 Antwerp, Belgium
| | - Valdir F Veiga-Junior
- Chemistry Department, Exact Sciences Institute, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Emerson Silva Lima
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| |
Collapse
|
27
|
Bernardes JR, Faria CC, Andrade IS, Ferreira ACF, Carvalho DP, Leitão AC, de Alencar TAM, Fortunato RS. Effect of the FE 2+ chelation by 2,2'-dipyridyl in the doxorubicin-induced lethality in breast tumor cell lines. Life Sci 2017; 192:128-135. [PMID: 29180001 DOI: 10.1016/j.lfs.2017.11.038] [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] [Received: 09/21/2017] [Revised: 11/17/2017] [Accepted: 11/23/2017] [Indexed: 12/22/2022]
Abstract
Breast cancer cells may exhibit changes in iron homeostasis, which results in increased labile iron pool (LIP) levels. Several studies highlight the crucial role of high LIP levels in the maintenance of tumor cell physiology. Iron chelators have been tested in anticancer therapy in combination with chemotherapeutic agents, to improve drug efficacy. Thus, the aim of this study was to evaluate the effect of 2,2'-dipyridyl (DIP), a Fe2+ chelator, in combination with doxorubicin (DOX) in breast tumor cells. The maximum concentration of DIP that did not significantly reduce the viability of MDA-MB-231 cells was 10μM and for MCF-7 cells was 50μM. We observed that MCF-7 had higher LIP levels than MDA-MB-231 cells. DIP alone increased ROS generation in MCF-7 cells, and DIP pretreatment reduced ROS generation induced by DOX treatment. In conclusion, the increase in MCF-7 cell viability induced by DIP pretreatment in DOX-treated cells seems to be related to an increase in the cellular antioxidant capacity and the iron chelator did not improve drug efficacy in the two breast tumor cell lines analyzed.
Collapse
Affiliation(s)
- Jéssica R Bernardes
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline C Faria
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iris S Andrade
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea Claudia Freitas Ferreira
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise P Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alvaro C Leitão
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana A M de Alencar
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S Fortunato
- Laboratório de Radiobiologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
28
|
Tavangar F, Sepehri H, Saghaeian Jazi M, Asadi J. Amphotericin B potentiates the anticancer activity of doxorubicin on the MCF-7 breast cancer cells. J Chem Biol 2017; 10:143-150. [PMID: 28685000 PMCID: PMC5480266 DOI: 10.1007/s12154-017-0172-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/02/2017] [Indexed: 01/05/2023] Open
Abstract
Despite the improvements in cancer treatment, breast cancer still remains the second most common cause of death from cancer in women. Doxorubicin (DOXO) is widely used for cancer treatment. However, drug resistance limits the treatment outcome. Here, we investigated the toxicity of DOXO in combination with an antifungal agent amphotericin B (AmB) against the MCF-7 breast cancer cell line. The cell viability was measured using MTT assay. The apoptosis was studied by caspase-8 and caspase-9 activity measurements and DNA fragmentation was investigated by TUNEL assay. The combination of two drugs significantly increased the apoptotic index and the caspase-8 and caspase-9 activities in comparison to DOXO-treated cells. Our finding showed that pre-treatment of MCF-7 cells with AmB synergistically exerted the anticancer effect of DOXO through the caspase-dependent apoptosis manner.
Collapse
Affiliation(s)
- Farzaneh Tavangar
- Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamid Sepehri
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Marie Saghaeian Jazi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Jahanbakhsh Asadi
- Biochemistry & Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
29
|
Mathematical and Computational Modeling in Complex Biological Systems. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5958321. [PMID: 28386558 PMCID: PMC5366773 DOI: 10.1155/2017/5958321] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 12/20/2016] [Accepted: 01/16/2017] [Indexed: 12/22/2022]
Abstract
The biological process and molecular functions involved in the cancer progression remain difficult to understand for biologists and clinical doctors. Recent developments in high-throughput technologies urge the systems biology to achieve more precise models for complex diseases. Computational and mathematical models are gradually being used to help us understand the omics data produced by high-throughput experimental techniques. The use of computational models in systems biology allows us to explore the pathogenesis of complex diseases, improve our understanding of the latent molecular mechanisms, and promote treatment strategy optimization and new drug discovery. Currently, it is urgent to bridge the gap between the developments of high-throughput technologies and systemic modeling of the biological process in cancer research. In this review, we firstly studied several typical mathematical modeling approaches of biological systems in different scales and deeply analyzed their characteristics, advantages, applications, and limitations. Next, three potential research directions in systems modeling were summarized. To conclude, this review provides an update of important solutions using computational modeling approaches in systems biology.
Collapse
|
30
|
Wang L, Chen Q, Qi H, Wang C, Wang C, Zhang J, Dong L. Doxorubicin-Induced Systemic Inflammation Is Driven by Upregulation of Toll-Like Receptor TLR4 and Endotoxin Leakage. Cancer Res 2016; 76:6631-6642. [PMID: 27680684 DOI: 10.1158/0008-5472.can-15-3034] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 08/10/2016] [Accepted: 09/06/2016] [Indexed: 11/16/2022]
Abstract
Doxorubicin is one of the most effective chemotherapeutic agents used for cancer treatment, but it causes systemic inflammation and serious multiorgan side effects in many patients. In this study, we report that upregulation of the proinflammatory Toll-like receptor TLR4 in macrophages by doxorubicin is an important step in generating its toxic side effects. In patient serum, doxorubicin treatment resulted in leakage of endotoxin and inflammatory cytokines into circulation. In mice, doxorubicin damaged the intestinal epithelium, which also resulted in leakage of endotoxin from the gut flora into circulation. Concurrently, doxorubicin increased TLR4 expression in macrophages both in vitro and in vivo, which further enhanced the sensitivity of these cells to endotoxin. Either depletion of gut microorganisms or blockage of TLR4 signaling effectively decreased doxorubicin-induced toxicity. Taken together, our findings suggest that doxorubicin-triggered leakage of endotoxin into the circulation, in tandem with enhanced TLR4 signaling, is a candidate mechanism underlying doxorubicin-induced systemic inflammation. Our study provides new insights for devising relevant strategies to minimize the adverse effects of chemotherapeutic agents such as doxorubicin, which may extend its clinical uses to eradicate cancer cells. Cancer Res; 76(22); 6631-42. ©2016 AACR.
Collapse
Affiliation(s)
- Lintao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China
| | - Haixia Qi
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR
| | - Cheng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China.,Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China.
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of life sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
31
|
Sorensen JC, Cheregi BD, Timpani CA, Nurgali K, Hayes A, Rybalka E. Mitochondria: Inadvertent targets in chemotherapy-induced skeletal muscle toxicity and wasting? Cancer Chemother Pharmacol 2016; 78:673-83. [PMID: 27167634 DOI: 10.1007/s00280-016-3045-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/20/2016] [Indexed: 12/19/2022]
Abstract
Chemotherapy has been associated with increased mitochondrial reactive oxygen species production, mitochondrial dysfunction and skeletal muscle atrophy leading to severe patient clinical complications including skeletal muscle fatigue, insulin resistance and wasting. The exact mechanisms behind this skeletal muscle toxicity are largely unknown, and as such co-therapies to attenuate chemotherapy-induced side effects are lacking. Here, we review the current literature describing the clinical manifestations and molecular origins of chemotherapy-induced myopathy with a focus on the mitochondria as the target organelle via which chemotherapeutic agents establish toxicity. We explore the likely mechanisms through which myopathy is induced, using the anthracycline doxorubicin, and the platinum-based alkylating agent oxaliplatin, as examples. Finally, we recommend directions for future research and outline the potential significance of these proposed directions.
Collapse
Affiliation(s)
- James C Sorensen
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia.,Australian Institute of Musculoskeletal Science, Western Health, Melbourne, 3021, Australia
| | - Beatrice D Cheregi
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia
| | - Cara A Timpani
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia.,Australian Institute of Musculoskeletal Science, Western Health, Melbourne, 3021, Australia
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia
| | - Alan Hayes
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia.,Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, 8001, Australia.,Australian Institute of Musculoskeletal Science, Western Health, Melbourne, 3021, Australia
| | - Emma Rybalka
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, 8001, Australia. .,Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, 8001, Australia. .,Australian Institute of Musculoskeletal Science, Western Health, Melbourne, 3021, Australia.
| |
Collapse
|
32
|
Dwivedi G, Gran MA, Bagchi P, Kemp ML. Dynamic Redox Regulation of IL-4 Signaling. PLoS Comput Biol 2015; 11:e1004582. [PMID: 26562652 PMCID: PMC4642971 DOI: 10.1371/journal.pcbi.1004582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/29/2015] [Indexed: 12/22/2022] Open
Abstract
Quantifying the magnitude and dynamics of protein oxidation during cell signaling is technically challenging. Computational modeling provides tractable, quantitative methods to test hypotheses of redox mechanisms that may be simultaneously operative during signal transduction. The interleukin-4 (IL-4) pathway, which has previously been reported to induce reactive oxygen species and oxidation of PTP1B, may be controlled by several other putative mechanisms of redox regulation; widespread proteomic thiol oxidation observed via 2D redox differential gel electrophoresis upon IL-4 treatment suggests more than one redox-sensitive protein implicated in this pathway. Through computational modeling and a model selection strategy that relied on characteristic STAT6 phosphorylation dynamics of IL-4 signaling, we identified reversible protein tyrosine phosphatase (PTP) oxidation as the primary redox regulatory mechanism in the pathway. A systems-level model of IL-4 signaling was developed that integrates synchronous pan-PTP oxidation with ROS-independent mechanisms. The model quantitatively predicts the dynamics of IL-4 signaling over a broad range of new redox conditions, offers novel hypotheses about regulation of JAK/STAT signaling, and provides a framework for interrogating putative mechanisms involving receptor-initiated oxidation. Incomplete reduction of oxygen during respiration results in the formation of highly reactive molecules known as reactive oxygen species (ROS) that react indiscriminately with cellular components and adversely affect cellular function. For a long time ROS were thought solely to be undesirable byproducts of respiration. Indeed, high levels of ROS are associated with a number of diseases. Despite these facts, antioxidants, agents that neutralize ROS, have not shown any clinical benefits when used as oral supplements. This paradox is partially explained by discoveries over the last two decades demonstrating that ROS are not always detrimental and may be essential for controlling physiological processes like cell signaling. However, the mechanisms by which ROS react with biomolecules are not well understood. In this work we have combined biological experiments with novel computational methods to identify the most important mechanisms of ROS-mediated regulation in the IL-4 signaling pathway of the immune system. We have also developed a detailed computer model of the IL-4 pathway and its regulation by ROS dependent and independent methods. Our work enhances the understanding of principles underlying regulation of cell signaling by ROS and has potential implications in advancing therapeutic methods targeting ROS and their adverse effects.
Collapse
Affiliation(s)
- Gaurav Dwivedi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Margaret A. Gran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Pritha Bagchi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Melissa L. Kemp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
33
|
Edwardson DW, Narendrula R, Chewchuk S, Mispel-Beyer K, Mapletoft JPJ, Parissenti AM. Role of Drug Metabolism in the Cytotoxicity and Clinical Efficacy of Anthracyclines. Curr Drug Metab 2015; 16:412-26. [PMID: 26321196 PMCID: PMC5398089 DOI: 10.2174/1389200216888150915112039] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 01/19/2023]
Abstract
Many clinical studies involving anti-tumor agents neglect to consider how these agents are metabolized within the host and whether the creation of specific metabolites alters drug therapeutic properties or toxic side effects. However, this is not the case for the anthracycline class of chemotherapy drugs. This review describes the various enzymes involved in the one electron (semi-quinone) or two electron (hydroxylation) reduction of anthracyclines, or in their reductive deglycosidation into deoxyaglycones. The effects of these reductions on drug antitumor efficacy and toxic side effects are also discussed. Current evidence suggests that the one electron reduction of anthracyclines augments both their tumor toxicity and their toxicity towards the host, in particular their cardiotoxicity. In contrast, the two electron reduction (hydroxylation) of anthracyclines strongly reduces their ability to kill tumor cells, while augmenting cardiotoxicity through their accumulation within cardiomyocytes and their direct effects on excitation/contraction coupling within the myocytes. The reductive deglycosidation of anthracyclines appears to inactivate the drug and only occurs under rare, anaerobic conditions. This knowledge has resulted in the identification of important new approaches to improve the therapeutic index of anthracyclines, in particular by inhibiting their cardiotoxicity. The true utility of these approaches in the management of cancer patients undergoing anthracycline-based chemotherapy remains unclear, although one such agent (the iron chelator dexrazoxane) has recently been approved for clinical use.
Collapse
Affiliation(s)
| | | | | | | | | | - Amadeo M Parissenti
- Dept. of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|
34
|
Systems biology approaches to enzyme kinetics: analyzing network models of drug metabolism. Methods Mol Biol 2014; 1113:317-34. [PMID: 24523119 DOI: 10.1007/978-1-62703-758-7_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Intracellular drug metabolism involves transport, bioactivation, conjugation, and other biochemical steps. The dynamics of these steps are each dependent on a number of other cellular factors that can ultimately lead to unexpected behavior. In this review, we discuss the confounding processes and coupled reactions within bioactivation networks that require a systems-level perspective in order to fully understand the time-varying behavior. When converting known in vitro characteristics of drug-enzyme interactions into descriptions of cellular systems, features such as substrate availability, cell-to-cell variability, and intracellular redox state deserve special focus. An example of doxorubicin bioactivation is used for discussing points of consideration when constructing and analyzing network models of drug metabolism.
Collapse
|
35
|
Singh S. Cytoprotective and regulatory functions of glutathione S-transferases in cancer cell proliferation and cell death. Cancer Chemother Pharmacol 2014; 75:1-15. [PMID: 25143300 DOI: 10.1007/s00280-014-2566-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/04/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE Glutathione S-transferases (GSTs) family of enzymes is best known for their cytoprotective role and their involvement in the development of anticancer drug resistance. Recently, emergence of non-detoxifying properties of GSTs has provided them with significant biological importance. Addressing the complex interactions of GSTs with regulatory kinases will help in understanding its precise role in tumor pathophysiology and in designing GST-centered anticancer strategies. METHODS We reviewed all published literature addressing the detoxification and regulatory roles of GSTs in the altered biology of cancer and evaluating novel agents targeting GSTs for cancer therapy. RESULTS The role of GSTs, especially glutathione S-transferase P1 isoform in tumoral drug resistance, has been the cause of intense debate. GSTs have been demonstrated to interact with different protein partners and modulate signaling pathways that control cell proliferation, differentiation and apoptosis. These specific functions of GSTs could lead to the development of new therapeutic approaches and to the identification of some interesting candidates for preclinical and clinical development. This review focuses on the crucial role played by GSTs in the development of resistance to anticancer agents and the major findings regarding the different modes of action of GSTs to regulate cell signaling.
Collapse
Affiliation(s)
- Simendra Singh
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Gautam Buddha Nagar, Greater Noida, UP, India,
| |
Collapse
|
36
|
Hua G, Liu Y, Li X, Xu P, Luo Y. Targeting glucose metabolism in chondrosarcoma cells enhances the sensitivity to doxorubicin through the inhibition of lactate dehydrogenase-A. Oncol Rep 2014; 31:2727-34. [PMID: 24789077 DOI: 10.3892/or.2014.3156] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 02/10/2014] [Indexed: 11/06/2022] Open
Abstract
Chondrosarcoma is a malignant cartilage-forming cancer composed of cells derived from transformed cells that produce cartilage. Conventional chemotherapy and radiotherapy have very limited efficacy in patients with advanced chondrosarcoma. In the present study, we reported a novel therapeutic approach in the treatment of chondrosarcoma cells. We detected that lactate dehydrogenase-A (LDHA) is highly active in chondrosarcoma cells and chondrosarcoma patient samples compared with normal chondrocyte cell lines and primary human chondrocyte. Moreover, chondrosarcoma cells exhibited elevated levels of LDHA expression under doxorubicin treatment. To further explore the mechanisms, we generated doxorubicin-resistant cells from SW1353 chondrosarcoma cell line. Notably, the activity and expression of LDHA are upregulated in doxorubicin-resistant cells. Moreover, our data showed a strong correlation between glucose metabolism and doxorubicin resistance in chondrosarcoma cells; doxorubicin-resistant cells displayed highly activated glucose metabolism and depended more on glucose supply. Finally, we reported a synergistic effect produced by incorporating doxorubicin with glycolysis inhibitors-oxamate in the combined treatment of chondrosarcoma cells in vitro and in vivo. In summary, the present study may aid in the development of new approaches using the combination of chemotherapeutic agents for the treatment of chondrosarcoma patients.
Collapse
Affiliation(s)
- Guojun Hua
- Department of Orthopaedics, The 101st Hospital of the People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Yunpeng Liu
- Department of Orthopaedics, The 101st Hospital of the People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Xiangyong Li
- Department of Hematology and Oncology, The 101st Hospital of the People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Peirong Xu
- Department of Orthopaedics, The 101st Hospital of the People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Yuchun Luo
- Department of Orthopaedics, The 101st Hospital of the People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| |
Collapse
|
37
|
Molecular analysis of the inhibitory effect of N-acetyl-L-cysteine on the proliferation and invasiveness of pancreatic cancer cells. Anticancer Drugs 2013; 24:504-18. [PMID: 23511429 DOI: 10.1097/cad.0b013e32836009d7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Preliminary studies have suggested that the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) may be effective in inhibiting the growth of pancreatic cancer cells. In-depth cellular and molecular analyses were carried out to determine NAC's mode of action in inhibiting the growth of a well-characterized pancreatic cancer cell line (AsPC-1). Standardized assays were used to monitor cellular growth, apoptosis, levels of ROS, cellular senescence, migration, and invasiveness. Cell stiffness was measured using atomic force microscopy. Gene expression was monitored by quantitative PCR. NAC significantly inhibits the growth and metastatic potential of AsPC-1 cells by inducing cell-cycle arrest in G1 and subsequent cellular senescence and decreased invasiveness. These anticancer properties are associated with an unexpected increase in the intracellular concentrations of ROS. NAC does not decrease the susceptibility of AsPC-1 cells to the anticancer drugs gemcitabine, mitomycin C, and doxorubicin. NAC-induced changes in gene expression are consistent with the onset of mesenchymal-to-epithelial transition. In conclusion, our findings indicate that NAC induces an integrated series of responses in AsPC-1 cells that make it a highly promising candidate for development as a pancreatic cancer therapeutic.
Collapse
|
38
|
Chitcholtan K, Sykes PH, Evans JJ. The resistance of intracellular mediators to doxorubicin and cisplatin are distinct in 3D and 2D endometrial cancer. J Transl Med 2012; 10:38. [PMID: 22394685 PMCID: PMC3316127 DOI: 10.1186/1479-5876-10-38] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 03/06/2012] [Indexed: 11/24/2022] Open
Abstract
Background Advanced endometrial cancer often shows resistance to clinical chemotherapy although potencies of anticancer drugs in vitro are promising. The disparity suggests that in vivo microenvironments are not recapitulated by in vitro models used for preclinical testing. However, spheroids replicate some important properties of tumours in vivo. Therefore, for the first time, we compared effects of doxorubicin and cisplatin on 3D multicellular structures and 2D cell monolayers of endometrial cancer cells. Methods 3D multicellular structures were generated by culturing cancer cells on non-adherent surfaces; and for comparison cell monolayers were cultured on adherent culture plates. Ishikawa, RL95-2, and KLE cell lines were studied. Morphologies of 3D multicellular structures were examined. After 48 hours treatment with anticancer drugs, apoptosis, proliferation, glucose metabolism and vascular endothelial growth factor (VEGF) were analysed. Immunostaining of PCNA, Glut-1, p-Erk1/2, SOD-1 and p-Akt1/2/3 was also performed. Results Distinct 3D multicellular morphologies were formed by three different endometrial cancer cell lines. Doxorubicin induced less apoptosis in 3D multicellular structures of high grade cancer cells (RL95-2 and KLE cell lines) than in cell monolayers. Parallel alterations in Erk1/2 phosphorylation and cell proliferation might suggest they were linked and again doxorubicin had less effect on 3D multicellular structures than cell monolayers. On the other hand, there was no correlation between altered glucose metabolism and proliferation. The responses depended on cancer cell lines and were apparently not mediated by altered Glut-1 levels. The level of SOD-1 was high in 3D cell cultures. The effects on VEGF secretion were various and cancer cell line dependent. Importantly, both doxorubicin and cisplatin had selective paradoxical stimulatory effects on VEGF secretion. The microenvironment within 3D multicellular structures sustained Akt phosphorylation, consistent with it having a role in anchorage-independent pathways. Conclusions The cancer cells responded to microenvironments in a distinctive manner. 3D multicellular structures exhibited greater resistance to the agents than 2D monolayers, and the differences between the culture formats were dependent on cancer cell lines. The effects of anticancer drugs on the intracellular mediators were not similar in 3D and 2D cultures. Therefore, using 3D cell models may have a significant impact on conclusions derived from screening drugs for endometrial carcinomas.
Collapse
Affiliation(s)
- Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | | | | |
Collapse
|
39
|
Finn NA, Kemp ML. Pro-oxidant and antioxidant effects of N-acetylcysteine regulate doxorubicin-induced NF-kappa B activity in leukemic cells. MOLECULAR BIOSYSTEMS 2012; 8:650-62. [PMID: 22134636 PMCID: PMC3337722 DOI: 10.1039/c1mb05315a] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Clinical debate has arisen over the consequences of antioxidant supplementation during cancer chemotherapy. While antioxidants may impede the efficacy of chemotherapy by scavenging reactive oxygen species and free radicals, it is also possible that antioxidants alleviate unwanted chemotherapy-induced toxicity, thus allowing for increased chemotherapy doses. These contradictory assertions suggest that antioxidant supplementation during chemotherapy treatment can have varied outcomes depending on the cellular context. To gain a more robust understanding of the role that antioxidants play in chemotherapy, we investigated the dose-dependent effects of the antioxidant, N-acetylcysteine (NAC), on the redox-mediated regulation of intracellular signaling. In this study, we systematically evaluated the effect of Dox-induced ROS on the NF-κB pathway in a pediatric acute lymphoblastic leukemia (ALL) cell line by measuring the thiol-based oxidative modifications of redox-sensitive proteins within the pathway. We report a functional consequence of NAC supplementation during doxorubicin (Dox) chemotherapy administration via the NF-kappa B (NF-κB) signal transduction pathway. The ability of NAC to alter Dox-induced NF-κB activity is contingent on the ROS-mediated S-glutathionylation of IKK-β. Moreover, the NAC-dependent alteration of intracellular glutathione redox balance, through pro-oxidant and antioxidant mechanisms, can be exploited to either promote or inhibit Dox-induced NF-κB activity in an NAC-concentration-dependent manner. We developed an electron-transfer-based computational model that predicts the effect of NAC pretreatment on Dox-induced NF-κB signaling for a range of NAC and Dox treatment combinations.
Collapse
Affiliation(s)
- Nnenna Adimora Finn
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30032-0363, USA; Fax: +1 404-894-4243; Tel: +1 404-385-6341
| | - Melissa Lambeth Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30032-0363, USA; Fax: +1 404-894-4243; Tel: +1 404-385-6341
| |
Collapse
|