1
|
Stolc V, Preto O, Karhanek M, Freund F, Griko Y, Loftus DJ, Ohayon MM. RNA-DNA Differences: Mechanisms, Oxidative Stress, Transcriptional Fidelity, and Health Implications. Antioxidants (Basel) 2025; 14:544. [PMID: 40427426 PMCID: PMC12108522 DOI: 10.3390/antiox14050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 05/29/2025] Open
Abstract
RNA-DNA differences (RDDs) challenge the traditional view of RNA as a faithful copy of DNA, arising through RNA editing, transcriptional errors, and oxidative damage. Reactive oxygen species (ROS) play a central role, inducing lesions like 8-oxo-guanine that compromise transcription and translation, leading to dysfunctional proteins. This review explores the biochemical basis of RDDs, their exacerbation under oxidative stress, and their dual roles in cellular adaptation and disease. RDDs contribute to genomic instability and are implicated in cancers, neurodegenerative disorders, and autoimmune diseases, while also driving phenotypic diversity. Drawing on terrestrial and spaceflight studies, we highlight the intersection of oxidative stress, RDD formation, and cellular dysfunction, proposing innovative mitigation approaches. Advancements in RDD detection and quantification, along with ROS management therapies, offer new avenues to restore cellular homeostasis and promote resilience. By positioning RDDs as a hallmark of genomic entropy, this review underscores the limits of biological adaptation. Furthermore, the prevalence of guanine-rich codons in antioxidant genes increases their susceptibility to ROS-induced oxidative lesions, linking redox stress, genomic instability, and constrained adaptation. These insights have profound implications for understanding aging, disease progression, and adaptive mechanisms in both terrestrial and space environments.
Collapse
Affiliation(s)
- Viktor Stolc
- NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Ondrej Preto
- Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Miloslav Karhanek
- Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | | | - Yuri Griko
- NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | | |
Collapse
|
2
|
Aloke C, Onisuru OO, Achilonu I. Glutathione S-transferase: A versatile and dynamic enzyme. Biochem Biophys Res Commun 2024; 734:150774. [PMID: 39366175 DOI: 10.1016/j.bbrc.2024.150774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
The dynamic and versatile group of enzymes referred to as glutathione S-transferases (GSTs) play diverse roles in cellular detoxification, safeguarding hosts from oxidative damage, and performing various other functions. This review explores different classes of GST, existence of polymorphisms in GST, functions of GST and utilizations of GST inhibitors in treatment of human diseases. The study indicates that the cytosolic GSTs, mitochondrial GSTs, microsomal GSTs, and bacterial proteins that provide resistance to Fosfomycin are the major classes. Given a GST, variation in its expression and function among individuals is due to the presence of polymorphic alleles that encode it. Genetic polymorphism might result in the modification of GST activity, thereby increasing individuals' vulnerability to harmful chemical compounds. GSTs have been demonstrated to play a regulatory function in cellular signalling pathways through kinases, S-Glutathionylation, and in detoxification processes. Various applications of bacterial GSTs and their potential roles in plants were examined. Targeting GSTs, especially GSTP1-1, is considered a potential therapeutic strategy for treating cancer and diseases linked to abnormal cell proliferation. Their role in cancer cell growth, differentiation, and resistance to anticancer agents makes them promising targets for drug development, offering prospects for the future.
Collapse
Affiliation(s)
- Chinyere Aloke
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa; Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Ebonyi State, Nigeria.
| | - Olalekan Olugbenga Onisuru
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| |
Collapse
|
3
|
Kang YT, Yang WJ, Huang HC, Tang SC, Ko JL. Exposure to nickel chloride induces epigenetic modification on detoxification enzyme glutathione S-transferase M2. ENVIRONMENTAL TOXICOLOGY 2024; 39:1729-1736. [PMID: 38050843 DOI: 10.1002/tox.24055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/23/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
Nickel (Ni) is a human carcinogen with genotoxic and epigenotoxic effects. Environmental and occupational exposure to Ni increases the risk of cancer and chronic inflammatory diseases. Our previous findings indicate that Ni alters gene expression through epigenetic regulation, specifically impacting E-cadherin and angiopoietin-like 4 (ANGPTL4), involved in epithelial-mesenchymal transition and migration. GST-M2, a member of the glutathione S-transferase (GST) enzyme family, plays a crucial role in cellular defense against oxidative damage and has been increasingly associated with cancer. GST-M2 overexpression inhibits lung cancer invasion and metastasis in vitro and in vivo. Hypermethylation of its promoter in cancer cells reduces gene expression, correlating with poor prognosis in non-small-cell lung cancer patients. The impact of Ni on GST-M2 remains unclear. We will investigate whether nickel exerts regulatory effects on GST-M2 through epigenetic modifications. Additionally, metformin, an antidiabetic drug, is being studied as a chemopreventive agent against nickel-induced damage. Our findings indicate that nickel chloride (NiCl2 ) exposure, both short-term and long-term, represses GST-M2 expression. However, the expression can be restored by demethylation agent 5-aza-2'-deoxycytidine and metformin. NiCl2 promotes hypermethylation of the GST-M2 promoter, as confirmed by methylation-specific PCR and bisulfite sequencing. Additionally, NiCl2 also influences histone acetylation, and metformin counteracts the suppressive effect of NiCl2 on histone H3 expression. Metformin reestablishes the binding of specificity protein 1 to the GST-M2 promoter, which is otherwise disrupted by NiCl2 . These findings elucidate the mechanism by which Ni reduces GST-M2 expression and transcriptional activity, potentially contributing to Ni-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Yu-Ting Kang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wan-Jung Yang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Genomic Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu Chih Huang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Sheau-Chung Tang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Khan A, Jahan F, Zahoor M, Ullah R, Albadrani GM, Mohamed HRH, Khisroon M. Association of genetic polymorphism of glutathione S-transferases with colorectal cancer susceptibility in snuff (Naswar) addicts. BRAZ J BIOL 2024; 84:e261509. [DOI: 10.1590/1519-6984.261509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
Abstract The current study aimed to investigate the relationship between polymorphisms in detoxifying (GSTM1, GSTT1, and GSTP1) genes and their association with colorectal cancer (CRC) in tobacco addicts of Pashtun ethnicity. Polymorphisms in the selected genes were genotyped in a case-control study consisting of 100 histologically confirmed male CRC patients and 100 birth-year and gender-matched healthy controls using the PCR−RFLP method. The GSTM1 null, and GSTT1 null genotypes were significantly contributed to the risk of CRC in the cases (OR= 3.131, 95% CI: 1.451−6.758, P = 0.004, and OR= 3.541, 95% CI: 1.716−7.306, P = 0.001, respectively), whereas the association observed for GSTP1 Val/Val (1.139, 95% CI: 0.356−3.644, P = 0.826) did not show statistical significance. The combined GSTM1 null and GSTT1 null showed a 41-fold increased risk (95% CI: 4.945−351.950, P = 0.001), while, the combined GSTM1 null and GSTP1 Ile/Val or Val/Val variant genotypes exhibited about 3-fold (95% CI: 1.196−7.414, P = 0.019) increased risk to CRC. Similarly, the combined GSTT1 null and GSTP1 Ile/Val or Val/Val variant genotypes showed about a 3-fold (95% CI: 1.285−8.101, P = 0.013) increased risk of CRC. In the combination of three GST genotypes, the GSTM1 null, GSTT1 null, and GSTP1 Ile/Val or Val/Val variant genotypes demonstrated a more than a 22-fold (95% CI: 2.441−212.106, P = 0.006) increased risk of CRC. Our findings suggest that GSTM1 and GSTT1 polymorphism and its combination with GSTP1 may be associated with CRC susceptibility in the Naswar addicted Pashtun population of Khyber Pakhtunkhwa, Pakistan.
Collapse
Affiliation(s)
- A. Khan
- University of Peshawar, Pakistan
| | - F. Jahan
- Shaheed Benazir Bhutto Women University Peshawar, Pakistan
| | | | - R. Ullah
- King Saud University, Saudi Arabia
| | | | | | | |
Collapse
|
5
|
Korczowska-Łącka I, Słowikowski B, Piekut T, Hurła M, Banaszek N, Szymanowicz O, Jagodziński PP, Kozubski W, Permoda-Pachuta A, Dorszewska J. Disorders of Endogenous and Exogenous Antioxidants in Neurological Diseases. Antioxidants (Basel) 2023; 12:1811. [PMID: 37891890 PMCID: PMC10604347 DOI: 10.3390/antiox12101811] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In diseases of the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), stroke, amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and even epilepsy and migraine, oxidative stress load commonly surpasses endogenous antioxidative capacity. While oxidative processes have been robustly implicated in the pathogenesis of these diseases, the significance of particular antioxidants, both endogenous and especially exogenous, in maintaining redox homeostasis requires further research. Among endogenous antioxidants, enzymes such as catalase, superoxide dismutase, and glutathione peroxidase are central to disabling free radicals, thereby preventing oxidative damage to cellular lipids, proteins, and nucleic acids. Whether supplementation with endogenously occurring antioxidant compounds such as melatonin and glutathione carries any benefit, however, remains equivocal. Similarly, while the health benefits of certain exogenous antioxidants, including ascorbic acid (vitamin C), carotenoids, polyphenols, sulforaphanes, and anthocyanins are commonly touted, their clinical efficacy and effectiveness in particular neurological disease contexts need to be more robustly defined. Here, we review the current literature on the cellular mechanisms mitigating oxidative stress and comment on the possible benefit of the most common exogenous antioxidants in diseases such as AD, PD, ALS, HD, stroke, epilepsy, and migraine. We selected common neurological diseases of a basically neurodegenerative nature.
Collapse
Affiliation(s)
- Izabela Korczowska-Łącka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Thomas Piekut
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Agnieszka Permoda-Pachuta
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| |
Collapse
|
6
|
The Upregulation of GSTO2 is Associated with Colon Cancer Progression and a Poor Prognosis. JOURNAL OF ONCOLOGY 2023; 2023:4931650. [PMID: 36688005 PMCID: PMC9848813 DOI: 10.1155/2023/4931650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023]
Abstract
Colorectal cancer is the second-leading cause of cancer-related mortality in the United States. Glutathione S-transferase can affect the development of cancer. Glutathione S-transferase omega 2, a member of the GST family, plays an important role in many tumors. However, the role of Glutathione S-transferase omega 2 in the development of colon cancer remains unclear. Herein, our study aimed to investigate the exact role of Glutathione S-transferase omega 2 in colon cancer. We used RNA sequencing data from The Cancer Genome Atlas and the Genotype-Tissue Expression database to analyze Glutathione S-transferase omega 2 expressions. Then, we explore the protein information of Glutathione S-transferase omega 2 in the Human Protein Atlas, GeneCards, and String database. In addition, western blot and immunohistochemistry were performed to evaluate the protein levels of Glutathione S-transferase omega 2 in colon cancer tissues. We acquire data from the Gene Expression Omnibus and The Cancer Genome Atlas databases. Also, we performed relevant prognostic analyses of these data. In addition, we performed a statistical analysis of the clinical data from The Cancer Genome Atlas database and the expression level of Glutathione S-transferase omega 2. Then, we performed Cox regression analysis and found independent risk factors for prognosis in patients with colon cancer. The Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analyses were used to explore the potential biological functions of Glutathione S-transferase omega 2. The infiltration of colon cancer-immune cells was evaluated by the CIBERSORT method. RNA silencing was performed using siRNA constructs in HCT-116 and HT-29 cell lines. Cell Counting Kit-8 and EdU assays were performed to determine cell proliferation. Transwell experiments and scratch tests were used to determine cell migration. As for the mRNA and protein expression levels of cells, we used quantitative real-time PCR and western blot to detect them. Our research shows that Glutathione S-transferase omega 2 is overexpressed in colon cancer patients, and this overexpression is associated with a poor prognosis. The high expression of Glutathione S-transferase omega 2 is significantly correlated stage with stage, M, and N classification progression in colon cancer by statistical analysis. Univariate and multivariate Cox regression analyses showed that Glutathione S-transferase omega 2 was an independent risk factor for poor prognosis in colon cancer. In addition, we also found that Glutathione S-transferase omega 2 expression levels can affect the immune microenvironment of colon cancer cells. Gene silencing of Glutathione S-transferase omega 2 in HT-29 and HCT-116 cells significantly inhibited tumor growth and migration. In summary, we found that Glutathione S-transferase omega 2 can be used as a molecular indicator of colon cancer prognosis. In vitro, gene silencing of Glutathione S-transferase omega 2 inhibited colon cancer cells' growth and migration.
Collapse
|
7
|
Kobzar O, Shulha Y, Buldenko V, Cherenok S, Silenko O, Kalchenko V, Vovk A. Inhibition of glutathione S-transferases by photoactive calix[4]arene α-ketophosphonic acids. Bioorg Med Chem Lett 2022; 77:129019. [DOI: 10.1016/j.bmcl.2022.129019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/20/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022]
|
8
|
Nanoparticle-based drug delivery systems to overcome gastric cancer drug resistance. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
9
|
Xu B, Tong T, Wang X, Liu F, Zhang X, Hu X, Li X, Yang X, Liao F. Short divalent ethacrynic amides as pro-inhibitors of glutathione S-transferase isozyme Mu and potent sensitisers of cisplatin-resistant ovarian cancers. J Enzyme Inhib Med Chem 2022; 37:728-742. [PMID: 35176963 PMCID: PMC8865112 DOI: 10.1080/14756366.2022.2038591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The linking of ethacrynic acid with ethylenediamine and 1,4-butanediamine gave EDEA and BDEA, respectively, as membrane-permeable divalent pro-inhibitors of glutathione S-transferase (GST). Their divalent glutathione conjugates showed subnanomolar inhibition and divalence-binding to GSTmu (GSTM) (PDB: 5HWL) at ∼0.35 min-1. In cisplatin-resistant SK-OV-3, COC1, SGC7901 and A549 cells, GSTM activities probed by 15 nM BDEA or EDEA revealed 5-fold and 1.0-fold increases in cisplatin-resistant SK-OV-3 and COC1 cells, respectively, in comparison with the susceptible parental cells. Being tolerable by HEK293 and LO2 cells, BDEA at 0.2 μM sensitised resistant SK-OV-3 and COC1 cells by ∼3- and ∼5-folds, respectively, released cytochrome c and increased apoptosis; EDEA at 1.0 μM sensitised resistant SK-OV-3 and A549 cells by ∼5- and ∼7-fold, respectively. EDEA at 1.7 μg/g sensitised resistant SK-OV-3 cells to cisplatin at 3.3 μg/g in nude mouse xenograft model. BDEA and EDEA are promising leads for probing cellular GSTM and sensitising cisplatin-resistant ovarian cancers.
Collapse
Affiliation(s)
- Bangtian Xu
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China.,Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Tingting Tong
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Fang Liu
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiang Zhang
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaolei Hu
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xinpeng Li
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaolan Yang
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Fei Liao
- Key Laboratory of Clinical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Franczyk B, Rysz J, Gluba-Brzózka A. Pharmacogenetics of Drugs Used in the Treatment of Cancers. Genes (Basel) 2022; 13:311. [PMID: 35205356 PMCID: PMC8871547 DOI: 10.3390/genes13020311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmacogenomics is based on the understanding of the individual differences in drug use, the response to drug therapy (efficacy and toxicity), and the mechanisms underlying variable drug responses. The identification of DNA variants which markedly contribute to inter-individual variations in drug responses would improve the efficacy of treatments and decrease the rate of the adverse side effects of drugs. This review focuses only on the impact of polymorphisms within drug-metabolizing enzymes on drug responses. Anticancer drugs usually have a very narrow therapeutic index; therefore, it is very important to use appropriate doses in order to achieve the maximum benefits without putting the patient at risk of life-threatening toxicities. However, the adjustment of the appropriate dose is not so easy, due to the inheritance of specific polymorphisms in the genes encoding the target proteins and drug-metabolizing enzymes. This review presents just a few examples of such polymorphisms and their impact on the response to therapy.
Collapse
Affiliation(s)
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
11
|
Phon BWS, Kamarudin MNA, Bhuvanendran S, Radhakrishnan AK. Transitioning pre-clinical glioblastoma models to clinical settings with biomarkers identified in 3D cell-based models: A systematic scoping review. Biomed Pharmacother 2022; 145:112396. [PMID: 34775238 DOI: 10.1016/j.biopha.2021.112396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/02/2022] Open
Abstract
Glioblastoma (GBM) remains incurable despite the overwhelming discovery of 2-dimensional (2D) cell-based potential therapeutics since the majority of them have met unsatisfactory results in animal and clinical settings. Incremental empirical evidence has laid the widespread need of transitioning 2D to 3-dimensional (3D) cultures that better mimic GBM's complex and heterogenic nature to allow better translation of pre-clinical results. This systematic scoping review analyses the transcriptomic data involving 3D models of GBM against 2D models from 22 studies identified from four databases (PubMed, ScienceDirect, Medline, and Embase). From a total of 499 genes reported in these studies, 313 (63%) genes were upregulated across 3D models cultured using different scaffolds. Our analysis showed that 4 of the replicable upregulated genes are associated with GBM stemness, epithelial to mesenchymal transition (EMT), hypoxia, and migration-related genes regardless of the type of scaffolds, displaying close resemblances to primitive undifferentiated tumour phenotypes that are associated with decreased overall survival and increased hazard ratio in GBM patients. The upregulation of drug response and drug efflux genes (e.g. cytochrome P450s and ABC transporters) mirrors the GBM genetic landscape that contributes to in vivo and clinical treatment resistance. These upregulated genes displayed strong protein-protein interactions when analysed using an online bioinformatics software (STRING). These findings reinforce the need for widespread transition to 3D GBM models as a relatively inexpensive humanised pre-clinical tool with suitable genetic biomarkers to bridge clinical gaps in potential therapeutic evaluations.
Collapse
Affiliation(s)
- Brandon Wee Siang Phon
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Muhamad N A Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia.
| | - Saatheeyavaane Bhuvanendran
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
12
|
Genetic polymorphism of glutathione S-transferases (GSTM1, GSTT1, and GSTP1) in patients with bullous pemphigoid in a Polish population. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction. Bullous pemphigoid (BP) is one of the most common bullous diseases with an autoimmune background. The etiology and pathogenesis of BP are believed to be influenced not only by environmental, genetic, and immunological factors as well as by oxidative stress. BP is observed more frequently in elderly patients. Additionally, more potent oxidative stress is observed just in old age. Glutathione S-transferases (GSTs) play key roles in the detoxification of xenobiotics, metabolism of endogenous substrates, and the defense against oxidative stress. The present study examines whether polymorphism of genes encoding three selected GSTs (GSTM1, GSTT1, and GSTP1) might be associated with a higher risk for BP.
Materials and methods. The study involved 71 patients with BP and 100 healthy volunteers from a Polish population. The presence of the deletion type polymorphism for GSTM1 and GSTT1 was confirmed by multiplex PCR. The Ile105Val GSTP1 polymorphism was analyzed by PCR-RFLP.
Results. It was observed that the combination of GSTM1 null/GSTT1 null/GSTP1 Ile/Val, Val/Val genotypes occurred more frequently in patients with BP (8.5%) than in controls (4.0%). The odds ratio for carriers of GSTM1 null/GSTT1 null/ GSTP1 Ile/Val, Val/Val genotypes was 2.22 (95% CI 0.60–8.16; p = 0.3727), but was not statistically significant.
Conclusions. The combination of GSTM1 null, GSTT1 null, GSTP1 Ile/Val, Val/Val genotypes might be related to a greater risk of BP in a Polish population. However, future studies including more individuals are required to confirm this.
Collapse
|
13
|
Usman M, Priya K, Pandit S, Gupta P. Cancer risk and nullity of Glutathione-S-transferase mu and theta 1 in occupational pesticide workers. Curr Pharm Biotechnol 2021; 23:932-945. [PMID: 34375184 DOI: 10.2174/1389201022666210810092342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/08/2022]
Abstract
Occupational exposure to pesticides has been associated with adverse health conditions, including genotoxicity and cancer. Nullity of GSTT1/GSTM1 increases the susceptibility of pesticide workers to these adverse health effects due to lack of efficient detoxification process created by the absence of these key xenobiotic metabolizing enzymes. However, this assertion does not seem to maintain its stance at all the time; some pesticide workers with the null genotypes do not present the susceptibility. This suggests the modulatory role of other confounding factors, genetic and environmental conditions. Pesticides, aggravated by the null GSTT1/GSTM1, cause genotoxicity and cancer through oxidative stress and miRNA dysregulation. Thus, the absence of these adverse health effects together with the presence of null GSTT1/GSTM1 genotypes demands further explanation. Also, understanding the mechanism behind the protection of cells - that are devoid of GSTT1/GSTM1 - from oxidative stress constitutes a great challenge and potential research area. Therefore, this review article highlights the recent advancements in the presence and absence of cancer risk in occupational pesticide workers with GSTT1 and GSTM1 null genotypes.
Collapse
Affiliation(s)
- Muhammad Usman
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, KP-III, Greater Noida- 201310 [U.P.], India
| | - Kanu Priya
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, KP-III, Greater Noida- 201310 [U.P.], India
| | - Soumya Pandit
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, KP-III, Greater Noida- 201310 [U.P.], India
| | - Piyush Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, KP-III, Greater Noida- 201310 [U.P.], India
| |
Collapse
|
14
|
Steel TR, Walsh F, Wieczorek-Błauż A, Hanif M, Hartinger CG. Monodentately-coordinated bioactive moieties in multimodal half-sandwich organoruthenium anticancer agents. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213890] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Acevedo-León D, Monzó-Beltrán L, Gómez-Abril SÁ, Estañ-Capell N, Camarasa-Lillo N, Pérez-Ebri ML, Escandón-Álvarez J, Alonso-Iglesias E, Santaolaria-Ayora ML, Carbonell-Moncho A, Ventura-Gayete J, Pla L, Martínez-Bisbal MC, Martínez-Máñez R, Bagán-Debón L, Viña-Almunia A, Martínez-Santamaría MA, Ruiz-Luque M, Alonso-Fernández J, Bañuls C, Sáez G. The Effectiveness of Glutathione Redox Status as a Possible Tumor Marker in Colorectal Cancer. Int J Mol Sci 2021; 22:ijms22126183. [PMID: 34201191 PMCID: PMC8226858 DOI: 10.3390/ijms22126183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
The role of oxidative stress (OS) in cancer is a matter of great interest due to the implication of reactive oxygen species (ROS) and their oxidation products in the initiation of tumorigenesis, its progression, and metastatic dissemination. Great efforts have been made to identify the mechanisms of ROS-induced carcinogenesis; however, the validation of OS byproducts as potential tumor markers (TMs) remains to be established. This interventional study included a total of 80 colorectal cancer (CRC) patients and 60 controls. By measuring reduced glutathione (GSH), its oxidized form (GSSG), and the glutathione redox state in terms of the GSSG/GSH ratio in the serum of CRC patients, we identified significant changes as compared to healthy subjects. These findings are compatible with the effectiveness of glutathione as a TM. The thiol redox state showed a significant increase towards oxidation in the CRC group and correlated significantly with both the tumor state and the clinical evolution. The sensitivity and specificity of serum glutathione levels are far above those of the classical TMs CEA and CA19.9. We conclude that the GSSG/GSH ratio is a simple assay which could be validated as a novel clinical TM for the diagnosis and monitoring of CRC.
Collapse
Affiliation(s)
- Delia Acevedo-León
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Lidia Monzó-Beltrán
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Odontotología-INCLIVA, Universidad de Valencia, 46010 Valencia, Spain; (L.M.-B.); (E.A.-I.)
| | - Segundo Ángel Gómez-Abril
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain;
| | - Nuria Estañ-Capell
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Natalia Camarasa-Lillo
- Servicio de Anatomía Patológica, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (N.C.-L.); (M.L.P.-E.); (J.E.-Á.)
| | - Marisa Luisa Pérez-Ebri
- Servicio de Anatomía Patológica, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (N.C.-L.); (M.L.P.-E.); (J.E.-Á.)
| | - Jorge Escandón-Álvarez
- Servicio de Anatomía Patológica, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (N.C.-L.); (M.L.P.-E.); (J.E.-Á.)
| | - Eulalia Alonso-Iglesias
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Odontotología-INCLIVA, Universidad de Valencia, 46010 Valencia, Spain; (L.M.-B.); (E.A.-I.)
| | - Marisa Luisa Santaolaria-Ayora
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Araceli Carbonell-Moncho
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Josep Ventura-Gayete
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Luis Pla
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València—Universitat de València, 46022 Valencia, Spain; (L.P.); (M.C.M.-B.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Maria Carmen Martínez-Bisbal
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València—Universitat de València, 46022 Valencia, Spain; (L.P.); (M.C.M.-B.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València—Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Departamento de Química, Universitat Politècnica de València, 46022 Valencia, Spain
- Departamento de Química Física, Universitat de València, Burjasot, 46100 Valencia, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València—Universitat de València, 46022 Valencia, Spain; (L.P.); (M.C.M.-B.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València—Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Departamento de Química, Universitat Politècnica de València, 46022 Valencia, Spain
- Departamento de Química Física, Universitat de València, Burjasot, 46100 Valencia, Spain
| | - Leticia Bagán-Debón
- Departamento de Estomatología, Facultad de Medicina y Odontología-INCLIVA, 46010 Valencia, Spain;
| | - Aurora Viña-Almunia
- Centro de Salud San Isidro, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - M. Amparo Martínez-Santamaría
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - María Ruiz-Luque
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Jorge Alonso-Fernández
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
| | - Celia Bañuls
- Servicio de Endocrinología y Nutrición, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain
- Correspondence: (C.B.); (G.S.); Tel.: +34-96-318-9132 (C.B.); +34-96-386-4160 (G.S.)
| | - Guillermo Sáez
- Servicio de Análisis Clínicos, Hospital Universitario Dr. Peset-FISABIO, 46017 Valencia, Spain; (D.A.-L.); (N.E.-C.); (M.L.S.-A.); (A.C.-M.); (J.V.-G.); (M.A.M.-S.); (M.R.-L.); (J.A.-F.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Odontotología-INCLIVA, Universidad de Valencia, 46010 Valencia, Spain; (L.M.-B.); (E.A.-I.)
- Correspondence: (C.B.); (G.S.); Tel.: +34-96-318-9132 (C.B.); +34-96-386-4160 (G.S.)
| |
Collapse
|
16
|
An updated meta-analysis showed smoking modify the association of GSTM1 null genotype on the risk of coronary heart disease. Biosci Rep 2021; 41:227694. [PMID: 33506866 PMCID: PMC7881159 DOI: 10.1042/bsr20200490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 12/06/2022] Open
Abstract
Background Oxidative stress is considered to be involved in the pathogenesis of coronary heart disease (CHD). Glutathione-S-transferase (GST) enzymes play important roles in antioxidant defenses and may influence CHD risk. The present meta-analysis was performed to investigate the link between glutathione S-transferase M1 (GSTM1) null genotype and CHD and to get a precise evaluation of interaction between GSTM1 null genotype and smoking by the case-only design. Methods PubMed and EMBASE databases were searched through 15 December 2020 to retrieve articles. Odds ratios (ORs) were pooled using either fixed-effects or random-effects models. Results Thirty-seven studies showed that GSTM1 null genotype was associated with risk of CHD in total population, Caucasians and Asians (for total population, OR = 1.38, 95% confidence interval (CI): 1.15, 1.65; for Caucasians, OR = 1.34, 95% CI: 1.04, 1.72; for Asians, OR = 1.40, 95% CI: 1.11, 1.77). After adjustment for heterogeneity, these relationships were still significant. After adjustment for heterogeneity, case-only analysis of 11 studies showed a positive multiplicative interaction between GSTM1 null genotype and smoking (ever smoking vs. never smoking) (OR = 1.27, 95% CI: 1.08, 1.50; I2 = 0%, P=0.553). Conclusions The overall results indicated that GSTM1 null genotype was associated with a higher risk of CHD, and the association may be affected by smoking status. This is the first meta-analysis to prove a positive effect of the interaction between GSTM1 null genotype and smoking status on the risk of CHD. Well-designed studies are needed to investigate the possible gene–gene or gene–environment interactions.
Collapse
|
17
|
Raju B, Choudhary S, Narendra G, Verma H, Silakari O. Molecular modeling approaches to address drug-metabolizing enzymes (DMEs) mediated chemoresistance: a review. Drug Metab Rev 2021; 53:45-75. [PMID: 33535824 DOI: 10.1080/03602532.2021.1874406] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Resistance against clinically approved anticancer drugs is the main roadblock in cancer treatment. Drug metabolizing enzymes (DMEs) that are capable of metabolizing a variety of xenobiotic get overexpressed in malignant cells, therefore, catalyzing drug inactivation. As evident from the literature reports, the levels of DMEs increase in cancer cells that ultimately lead to drug inactivation followed by drug resistance. To puzzle out this issue, several strategies inclusive of analog designing, prodrug designing, and inhibitor designing have been forged. On that front, the implementation of computational tools can be considered a fascinating approach to address the problem of chemoresistance. Various research groups have adopted different molecular modeling tools for the investigation of DMEs mediated toxicity problems. However, the utilization of these in-silico tools in maneuvering the DME mediated chemoresistance is least considered and yet to be explored. These tools can be employed in the designing of such chemotherapeutic agents that are devoid of the resistance problem. The current review canvasses various molecular modeling approaches that can be implemented to address this issue. Special focus was laid on the development of specific inhibitors of DMEs. Additionally, the strategies to bypass the DMEs mediated drug metabolism were also contemplated in this report that includes analogs and pro-drugs designing. Different strategies discussed in the review will be beneficial in designing novel chemotherapeutic agents that depreciate the resistance problem.
Collapse
Affiliation(s)
- Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
18
|
Pamidimukkala K, Rani Inala M. Amalgamation of quercetin with anastrozole and capecitabine: A novel combination to treat breast and colon cancers – An in vitro study. J Cancer Res Ther 2021; 19:S93-S105. [PMID: 37147989 DOI: 10.4103/jcrt.jcrt_599_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Context Globally, cancer stands as the principle cause of mortality and immediate attention on its treatment options is required. Natural compounds stay at first priority in encountering novel therapeutics without adverse effects. Aim The aim of the study is to extract flavonol quercetin from leafy vegetables of Anethum graveolens L. and Raphanus sativus L. and find out its potential in combination with drugs used for chemotherapy to reduce the adverse effects of drugs. Settings and Design Observational study. Materials And Methods Column chromatography is used for quercetin extraction and anticancer activity of quercetin + anastrozole and quercetin + capecitabine were determined by (4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay (MTT), apoptosis assay, cell cycle analysis, mitochondrial membrane potential, and caspase 3 expression. Statistical Analysis Used Cytotoxic assay results were assessed by mean, standard deviation and ANOVA; and results were compared for determining its significance. Results The results noted that quercetin at very less concentration (16 and 31 μg/ml on Michigan Cancer Foundation-7 and 43 and 46 μg/ml on COLO 320) in combination with anastrozole and capecitabine was able to control the growth of cells, increase cell death, arrest cell cycle, and induce mitochondrial depolarization and expression of caspase 3. Conclusions The natural compound used in the present study is effective in treating breast and colon cancer at minimal concentrations in combination with the drugs. This combinational treatment appears to be reported for the first time in the present study.
Collapse
|
19
|
Yamashita Y, Sakakibara H, Toda T, Ashida H. Insights into the potential benefits of black soybean ( Glycine max L.) polyphenols in lifestyle diseases. Food Funct 2020; 11:7321-7339. [PMID: 32852022 DOI: 10.1039/d0fo01092h] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Black soybean (Glycine max L.), a cultivar containing abundant polyphenols in its seed coat such as anthocyanins and flavan-3-ols, has been reported to possess various health benefits toward lifestyle diseases. In this review article, the safety evaluation of polyphenol-rich black soybean seed coat extract (BE) and absorption of BE polyphenols are summarized. Additionally, we describe the antioxidant activity of BE polyphenols and their ability to induce antioxidant enzymes. The health benefits of BE and its polyphenols, such as anti-obesity and anti-hyperglycemic activities through the activation of AMP-activated protein kinase and translocation of glucose transporter 4, respectively, are also discussed. Furthermore, we found that black soybean polyphenols were involved in the improvement of vascular function. These emerging data require further investigation in scientific studies and human trials to evaluate the prevention of lifestyle diseases using black soybean polyphenols.
Collapse
Affiliation(s)
- Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | | | - Toshiya Toda
- Department of Innovative Food Sciences, School of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya 663-8558, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
20
|
Branco V, Pimentel J, Brito MA, Carvalho C. Thioredoxin, Glutathione and Related Molecules in Tumors of the Nervous System. Curr Med Chem 2020; 27:1878-1900. [PMID: 30706774 DOI: 10.2174/0929867326666190201113004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 09/14/2018] [Accepted: 11/28/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Central Nervous System (CNS) tumors have a poor survival prognosis due to their invasive and heterogeneous nature, in addition to the resistance to multiple treatments. OBJECTIVE In this paper, the main aspects of brain tumor biology and pathogenesis are reviewed both for primary tumors of the brain, (i.e., gliomas) and for metastasis from other malignant tumors, namely lung cancer, breast cancer and malignant melanoma which account for a high percentage of overall malignant brain tumors. We review the role of antioxidant systems, namely the thioredoxin and glutathione systems, in the genesis and/or progression of brain tumors. METHODS Although overexpression of Thioredoxin Reductase (TrxR) and Thioredoxin (Trx) is often linked to increased malignancy rate of brain tumors, and higher expression of Glutathione (GSH) and Glutathione S-Transferases (GST) are associated to resistance to therapy, several knowledge gaps still exist regarding for example, the role of Peroxiredoxins (Prx), and Glutaredoxins (Grx). CONCLUSION Due to their central role in redox homeostasis and ROS scavenging, redox systems are potential targets for new antitumorals and examples of innovative therapeutics aiming at improving success rates in brain tumor treatment are discussed.
Collapse
Affiliation(s)
- Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Pimentel
- Laboratory of Neuropathology, Department of Neurology, Hospital de Santa Maria (CHLN), Av. Prof. Egas Moniz, 1649-036 Lisboa, Portugal.,Faculty of Medicine, Lisbon University, Av. Prof. Egas Moniz, 1649-036 Lisboa, Portugal
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
21
|
Hattinger CM, Patrizio MP, Luppi S, Serra M. Pharmacogenomics and Pharmacogenetics in Osteosarcoma: Translational Studies and Clinical Impact. Int J Mol Sci 2020; 21:E4659. [PMID: 32629971 PMCID: PMC7369799 DOI: 10.3390/ijms21134659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
High-grade osteosarcoma (HGOS) is a very aggressive bone tumor which primarily affects adolescents and young adults. Although not advanced as is the case for other cancers, pharmacogenetic and pharmacogenomic studies applied to HGOS have been providing hope for an improved understanding of the biology and the identification of genetic biomarkers, which may impact on clinical care management. Recent developments of pharmacogenetics and pharmacogenomics in HGOS are expected to: i) highlight genetic events that trigger oncogenesis or which may act as drivers of disease; ii) validate research models that best predict clinical behavior; and iii) indicate genetic biomarkers associated with clinical outcome (in terms of treatment response, survival probability and susceptibility to chemotherapy-related toxicities). The generated body of information may be translated to clinical settings, in order to improve both effectiveness and safety of conventional chemotherapy trials as well as to indicate new tailored treatment strategies. Here, we review and summarize the current scientific evidence for each of the aforementioned issues in view of possible clinical applications.
Collapse
Affiliation(s)
| | | | | | - Massimo Serra
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, 40136 Bologna, Italy; (C.M.H.); (M.P.P.); (S.L.)
| |
Collapse
|
22
|
Chaicharoenaudomrung N, Kunhorm P, Promjantuek W, Rujanapun N, Heebkaew N, Soraksa N, Noisa P. Transcriptomic Profiling of 3D Glioblastoma Tumoroids for the Identification of Mechanisms Involved in Anticancer Drug Resistance. In Vivo 2020; 34:199-211. [PMID: 31882480 DOI: 10.21873/invivo.11762] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Among various types of brain tumors, glioblastoma is the most malignant and highly aggressive brain tumor that possesses a high resistance against anticancer drugs. To understand the underlined mechanisms of tumor drug resistance, a new and more effective research approach is required. The three dimensional (3D) in vitro cell culture models could be a potential approach to study cancer features and biology, as well as screen for anti-cancer agents due to the close mimicry of the 3D tumor microenvironments. MATERIALS AND METHODS With our developed 3D alginate scaffolds, Ilumina RNA-sequencing was used to transcriptomically analyze and compare the gene expression profiles between glioblastoma cells in traditional 2-dimensional (2D) monolayer and in 3D Ca-alginate scaffolds at day 14. To verify the reliability and accuracy of Illumina RNA-Sequencing data, ATP-binding cassette transporter genes were chosen for quantitative real-time polymerase chain reaction) verification. RESULTS The results showed that 7,411 and 3,915 genes of the 3D glioblastoma were up-regulated and down-regulated, respectively, compared with the 2D-cultured glioblastoma. Furthermore, the Kyoto Encyclopaedia of Genes and Genomes pathway analysis revealed that genes related to the cell cycle and DNA replication were enriched in the group of down-regulated gene. On the other hand, the genes involved in mitogen-activated protein kinase signaling, autophagy, drug metabolism through cytochrome P450, and ATP-binding cassette transporter were found in the up-regulated gene collection. CONCLUSION 3D glioblastoma tumoroids might potentially serve as a powerful platform for exploring glioblastoma biology. They can also be valuable in anti-glioblastoma drug screening, as well as the identification of novel molecular targets in clinical treatment of human glioblastoma.
Collapse
Affiliation(s)
- Nipha Chaicharoenaudomrung
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Phongsakorn Kunhorm
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Wilasinee Promjantuek
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Narawadee Rujanapun
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Nudjanad Heebkaew
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Natchadaporn Soraksa
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Parinya Noisa
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| |
Collapse
|
23
|
Aksoy M, Karaman M, Güller P, Güller U, Küfrevioğlu Öİ. In Vitro Inhibition Effect and Molecular Docking Study of Curcumin, Resveratrol, and Quercetin on Human Erythrocyte Glutathione Transferase. ACTA ACUST UNITED AC 2020. [DOI: 10.2174/1573408016666191231123544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background:Chemotherapy has shown varying success rates in the treatment of metastatic cancer in the last 50 years. One of the problems in the use of many chemotherapeutic agents is to increase the expression of glutathione transferase enzyme (GST; EC 2.5.1.18). Therefore, the development of GST inhibitors is important to improve the effectiveness of antitumor drugs and to overcome multi-drug resistance.Introduction:Glutathione S-transferases (GSTs) are a major member of enzymes serving in the detoxification of exogenous and endogenous substances. But, it has been reported that GSTs are overexpressed in many tumour cells, and it has been found to be related to developing resistance to anticancer drugs by these cells. The development of GST inhibitors is important to increase the efficacy of antitumor drugs and overcome multi-drug resistance. The aim of our study was to investigate the effect of natural compounds including curcumin, resveratrol, and quercetin on GST enzyme activity. We also aimed to specify inhibition mechanism of the compounds on human erythrocytes GST (hGST) with in silico study.Method:GST was purified from human erythrocytes using affinity chromatography (glutathione agarose). The enzyme purity was checked with SDS-PAGE. After the inhibitory effect of the curcumin, quercetin, resveratrol was investigated. Lastly, inhibition mechanisms of these natural compound were identified with induced-fit docking method.Result:GST was purified with 19.31% yield from human erythrocytes. In inhibition studies, Ki values of curcumin, quercetin, resveratrol were determined as 0.0021 ± 0.0008, 0.0257 ± 0.0011, 663.3301 ± 0.0936 µM respectively. According to our results, all natural products showed the inhibition effect and the order of inhibition is as follows: curcumin ˃ quercetin ˃ resveratrol.Conclusion:According to the results of the in vitro and in silico studies, it can be said that curcumin, quercetin, resveratrol are the inhibitors of human erythrocyte GST. In conclusion, these observations may be of great importance for the potential use of these natural compounds as chemopreventive agents.
Collapse
Affiliation(s)
- Mine Aksoy
- Department of Chemistry, Faculty of Science, Atatürk University, 25240-Erzurum, Turkey
| | - Muhammet Karaman
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Kilis 7 Aralik University, 79000 Kilis, Turkey
| | - Pınar Güller
- Department of Chemistry, Faculty of Science, Atatürk University, 25240-Erzurum, Turkey
| | - Uğur Güller
- Department of Food Engineering, Faculty of Engineering, Igdir University, 76100-Igdir, Turkey
| | - Ö. İrfan Küfrevioğlu
- Department of Chemistry, Faculty of Science, Atatürk University, 25240-Erzurum, Turkey
| |
Collapse
|
24
|
Pljesa-Ercegovac M, Savic-Radojevic A, Coric V, Radic T, Simic T. Glutathione transferase genotypes may serve as determinants of risk and prognosis in renal cell carcinoma. Biofactors 2020; 46:229-238. [PMID: 31483924 DOI: 10.1002/biof.1560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/18/2019] [Indexed: 12/25/2022]
Abstract
Renal cell carcinoma (RCC) represents a group of histologically similar neoplasms with significant intratumor and intertumor genetic heterogeneity. Recognized risk factors for RCC development include smoking, hypertension, obesity, as well as von Hippel-Lindau (VHL) disease. Inactivation of VHL, deregulated nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway, and altered redox homeostasis, together with changes in glutathione transferase (GST) profile, are considered as important contributing factors in RCC development and progression. Although the available results of both gene-gene and gene-environment analysis are quite heterogeneous, they clearly indicate that certain GST genotypes may play a role as risk modifiers, either individually or in combination with other Phase I or Phase II gene polymorphisms, as well as in subjects exposed to relevant substrates. Seemingly, GST genotyping could identify individuals with impaired detoxification in renal parenchyma that are at higher risk of developing RCC. In addition to well established roles of GSTs in conjugation and biotransformation of xenobiotics, GSTs have emerged as significant regulators of pathways determining cell proliferation and survival. Indeed, there are evidence in favor of GST significance, not only in terms of risk for RCC development, but also with respect to progression and prognosis. So far, GSTM1-active genotype was confirmed to be an independent predictor of higher risk of overall mortality. Therefore, it is reasonable to assume that certain GST variants may assist in individual RCC risk assessment, as well as postoperative prognosis. Even more, GST profiling might contribute to development of personalized targeted therapy in RCC patients.
Collapse
Affiliation(s)
- Marija Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tanja Radic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
25
|
Ruan T, Liu W, Tao K, Wu C. A Review of Research Progress in Multidrug-Resistance Mechanisms in Gastric Cancer. Onco Targets Ther 2020; 13:1797-1807. [PMID: 32184615 PMCID: PMC7053652 DOI: 10.2147/ott.s239336] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/15/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is one of the most common malignant tumors, and it is also one of the leading causes of cancer death worldwide. Because of its insidious symptoms and lack of early dictation screening, many cases of gastric cancer are at late stages which make it more complicated to cure. For these advanced-stage gastric cancers, combination therapy of surgery, chemotherapy, radiotherapy and target therapy would bring more benefit to the patients. However, the drug-resistance to the chemotherapy restricts its effect and might lead to treatment failure. In this review article, we discuss the mechanisms which have been found in recent years of drug resistance in gastric cancer. And we also want to find new approaches to counteract chemotherapy resistance and bring more benefits to the patients.
Collapse
Affiliation(s)
- Tuo Ruan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuanqing Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
26
|
Narayanankutty A, Job JT, Narayanankutty V. Glutathione, an Antioxidant Tripeptide: Dual Roles in Carcinogenesis and Chemoprevention. Curr Protein Pept Sci 2020; 20:907-917. [PMID: 30727890 DOI: 10.2174/1389203720666190206130003] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 12/12/2022]
Abstract
Glutathione (GSH or reduced glutathione) is a tripeptide of gamma-Glutamyl-cysteinylglycine and the predominant intracellular antioxidant in many organisms including humans. GSH and associated enzymes are controlled by a transcription factor-nuclear factor-2 related erythroid factor-2 (Nrf2). In cellular milieu, GSH protects the cells essentially against a wide variety of free radicals including reactive oxygen species, lipid hydroperoxides, xenobiotic toxicants, and heavy metals. It has two forms, the reduced form or reduced glutathione (GSH) and oxidized form (GSSG), where two GSH moieties combine by sulfhydryl bonds. Glutathione peroxidase (GPx) and glutathione-s-transferase (GST) essentially perform the detoxification reactions using GSH, converting it into GSSG. Glutathione reductase (GR) operates the salvage pathway by converting GSSG to GSH with the expense of NADPH and restores the cellular GSH pool. Hence, GSH and GSH-dependent enzymes are necessary for maintaining the normal redox balance in the body and help in cell survival under stress conditions. In addition, GST removes various carcinogenic compounds offering a chemopreventive property, whereas the GSH system plays a significant role in regulating the cellular survival by offering redox stability in a variety of cancers including prostate, lung, breast, and colon cancer. Studies have also indicated that GSH inhibitors, such as buthionine sulfoximine, improve the chemo-sensitivity in cancer cells. In addition, GSH and dependent enzymes provide a survival advantage for cancer cells against chemotherapeutic drugs and radiotherapy.
Collapse
Affiliation(s)
- Arunaksharan Narayanankutty
- Postgraduate & Research Department of Zoology, St. Joseph's College (Autonomous), Devagiri (Affiliated to University of Calicut), Calicut- 673 019, Kerala, India
| | - Joice Tom Job
- Postgraduate & Research Department of Zoology, St. Joseph's College (Autonomous), Devagiri (Affiliated to University of Calicut), Calicut- 673 019, Kerala, India
| | | |
Collapse
|
27
|
Abstract
The mercapturic acid pathway is a major route for the biotransformation of xenobiotic and endobiotic electrophilic compounds and their metabolites. Mercapturic acids (N-acetyl-l-cysteine S-conjugates) are formed by the sequential action of the glutathione transferases, γ-glutamyltransferases, dipeptidases, and cysteine S-conjugate N-acetyltransferase to yield glutathione S-conjugates, l-cysteinylglycine S-conjugates, l-cysteine S-conjugates, and mercapturic acids; these metabolites constitute a "mercapturomic" profile. Aminoacylases catalyze the hydrolysis of mercapturic acids to form cysteine S-conjugates. Several renal transport systems facilitate the urinary elimination of mercapturic acids; urinary mercapturic acids may serve as biomarkers for exposure to chemicals. Although mercapturic acid formation and elimination is a detoxication reaction, l-cysteine S-conjugates may undergo bioactivation by cysteine S-conjugate β-lyase. Moreover, some l-cysteine S-conjugates, particularly l-cysteinyl-leukotrienes, exert significant pathophysiological effects. Finally, some enzymes of the mercapturic acid pathway are described as the so-called "moonlighting proteins," catalytic proteins that exert multiple biochemical or biophysical functions apart from catalysis.
Collapse
Affiliation(s)
- Patrick E Hanna
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
28
|
Banerjee BD, Kumar R, Thamineni KL, Shah H, Thakur GK, Sharma T. Effect of Environmental Exposure and Pharmacogenomics on Drug Metabolism. Curr Drug Metab 2020; 20:1103-1113. [PMID: 31933442 DOI: 10.2174/1389200221666200110153304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/02/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pesticides are major xenobiotic compounds and environmental pollutants, which are able to alter drug-metabolizing enzyme as well as pharmacokinetics of drugs. Subsequent to the release of the human genome project, genetic variations (polymorphism) become an integral part of drug development due to their influence on disease susceptibility/ progression of the disease and their impact on drug absorption, distribution, metabolism of active metabolites and finally excretion of the drug. Genetic polymorphisms crucially regulate pharmacokinetics and pharmacodynamics of drugs under the influence of physiological condition, lifestyle, as well as pathological conditions collectively. OBJECTIVE To review all the evidence concerning the effect of environmental exposure on drug metabolism with reference to pharmacogenomics. METHODS Scientific data search and review of basic, epidemiological, pharmacogenomics and pharmacokinetics studies were undertaken to evaluate the influence of environmental contaminants on drug metabolism. RESULTS Various environmental contaminants like pesticides effectively alter drug metabolism at various levels under the influence of pharmacogenomics, which interferes with pharmacokinetics of drug metabolism. Genetic polymorphism of phase I and phase II xenobiotic-metabolizing enzymes remarkably alters disease susceptibility as well as the progression of disease under the influence of various environmental contaminants at various levels. CONCLUSION Individual specific drug response may be attributed to a large variety of factors alone or in combination ranging from genetic variations (SNP, insertion, deletion, duplication etc.) to physiological setting (gender, age, body size, and ethnicity), environmental or lifestyle factors (radiation exposure, smoking, alcohol, nutrition, exposure to toxins, etc.); and pathological conditions (obesity, diabetes, liver and renal function).
Collapse
Affiliation(s)
- Basu Dev Banerjee
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| | - Ranjeet Kumar
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| | - Krishna Latha Thamineni
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| | - Harendra Shah
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| | - Gaurav Kumar Thakur
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| | - Tusha Sharma
- Environmental Biochemistry and Molecular Biology Laboratory, Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Dilshad Garden, Delhi-110095, India
| |
Collapse
|
29
|
Zhang M, Wu SQ, He JQ. Are genetic variations in glutathione S-transferases involved in anti-tuberculosis drug-induced liver injury? A meta-analysis. J Clin Pharm Ther 2019; 44:844-857. [PMID: 31378997 DOI: 10.1111/jcpt.13006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/20/2019] [Accepted: 07/05/2019] [Indexed: 02/05/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE As a crucial protective role in the detoxifying mechanisms of drugs, glutathione S-transferases (GSTs) may affect an individual patient's susceptibility to anti-tuberculosis drug-induced liver injury (ATLI). However, the results of studies investigate the association between GSTM1, GSTT1 and GSTP1 polymorphisms and risk of ATLI are inconclusive. A meta-analysis on this topic was performed. METHODS PubMed, EMBASE, ISI web of science and the Chinese National Knowledge Infrastructure (CNKI) were systematically searched to identify relevant studies. Odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were calculated. Heterogeneity among articles and publication bias were also tested. RESULTS AND DISCUSSION After excluding one study as an outlier, the null GSTM1 genotype was associated with an increased risk of ATLI (OR = 1.270, 95% CI (1.014-1.590, P = .038), especially in East Asians (OR = 1.501, 95% CI (1.303-1.730). With similar exclusion, the null GSTT1 genotype increased the risk of ATLI in the total population (OR = 1.169, 95% CI: 1.028-1.330) and in Indians (OR = 1.732, 95% CI: 1.229-2.416). No statistically significant association was observed between the mutant GSTP1 genotype with risk of ATLI, which may need more rigorous and uniform case-control or cohort studies for more robust inferences. WHAT IS NEW AND CONCLUSION This up-to-date meta-analysis strongly suggests associations of GSTM1 and GSTT1 polymorphisms with ATLI. The results show the increased risk of ATL1 with the null GSTM1 and GSTT1 genotype on ATLI development. No such association is shown with the mutant GSTP1 genotype.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Shou-Quan Wu
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jian-Qing He
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Capelôa T, Benyahia Z, Zampieri LX, Blackman MCNM, Sonveaux P. Metabolic and non-metabolic pathways that control cancer resistance to anthracyclines. Semin Cell Dev Biol 2019; 98:181-191. [PMID: 31112797 DOI: 10.1016/j.semcdb.2019.05.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Anthracyclines Doxorubicin, Epirubicin, Daunorubicin and Idarubicin are used to treat a variety of tumor types in the clinics, either alone or, most often, in combination therapies. While their cardiotoxicity is well known, the emergence of chemoresistance is also a major issue accounting for treatment discontinuation. Resistance to anthracyclines is associated to the acquisition of multidrug resistance conferred by overexpression of permeability glycoprotein-1 or other efflux pumps, by altered DNA repair, changes in topoisomerase II activity, cancer stemness and metabolic adaptations. This review further details the metabolic aspects of resistance to anthracyclines, emphasizing the contributions of glycolysis, the pentose phosphate pathway and nucleotide biosynthesis, glutathione, lipid metabolism and autophagy to the chemoresistant phenotype.
Collapse
Affiliation(s)
- Tânia Capelôa
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Zohra Benyahia
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Luca X Zampieri
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marine C N M Blackman
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
31
|
Sun C, Gu Y, Chen G, Du Y. Bioinformatics Analysis of Stromal Molecular Signatures Associated with Breast and Prostate Cancer. J Comput Biol 2019; 26:1130-1139. [PMID: 31180245 DOI: 10.1089/cmb.2019.0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This study aimed to identify stromal molecular signatures associated with breast and prostate cancer. The microarray data GSE26910 was downloaded from Gene Expression Omnibus database, including six invasive breast tumor stroma, six matched normal controls, six invasive prostate tumor stroma, and six matched controls. The differentially expressed genes (DEGs) in invasive breast and prostate tumors stroma were, respectively, identified. Then common stromal genes (B_P.DEGs) were further screened. Protein-protein interaction (PPI) network was constructed and Gene Ontology analysis was performed. Besides, gene-chemical interactions were mapped in Comparative Toxicogenomics Database to screen the chemicals related to feature genes. The results showed that, in total, 16 B_P.DEGs were identified. Thereinto, only seven B_P.DEGs were mapped into PPI, and only four functional modules (adenylate cyclase activating polypeptide 1 (pituitary) receptor type I (ADCYAP1R1) module, aspartoacylase (ASPA) module, glutathione S-transferase mu 5 (GSTM5) module, and periplakin (PPL) module) were involved in important biological processes associated with cancer progression. In addition, the chemicals, such as dihydrotestosterone, apocarotenal, testosterone, and progesterone, were screened for the roles of feature genes in the progression of breast and prostate cancer. In conclusion, ADCYAP1R1, GSTM5, and PPL were stromal molecular signatures and might play a key role in the progression of breast and prostate cancer.
Collapse
Affiliation(s)
- Chao Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Gu
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoqing Chen
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yibao Du
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Chiarella P, Capone P, Carbonari D, Sisto R. A Predictive Model Assessing Genetic Susceptibility Risk at Workplace. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16112012. [PMID: 31195756 PMCID: PMC6603935 DOI: 10.3390/ijerph16112012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 01/08/2023]
Abstract
(1) Background: The study of susceptibility biomarkers in the immigrant workforce integrated into the social tissue of European host countries is always a challenge, due to high individual heterogeneity and the admixing of different ethnicities in the same workplace. These workers having distinct cultural backgrounds, beliefs, diets, and habits, as well as a poor knowledge of the foreign language, may feel reluctant to donate their biological specimens for the biomonitoring research studies. (2) Methods: A model predicting ethnicity-specific susceptibility based on principal component analysis has been conceived, using the genotype frequency of the investigated populations available in publicly accessible databases. (3) Results: Correlations among ethnicities and between ethnic and polymorphic genes have been found, and low/high-risk profiles have been identified as valuable susceptibility biomarkers. (4) Conclusions: In the absence of workers’ consent or access to blood genotyping, ethnicity represents a good indicator of the subject’s genotype. This model, associating ethnicity-specific genotype frequency with the susceptibility biomarkers involved in the metabolism of toxicants, may replace genotyping, ensuring the necessary safety and health conditions of workers assigned to hazardous jobs.
Collapse
Affiliation(s)
- Pieranna Chiarella
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Via Fontana Candida 1, 00078 Monteporzio Catone, Rome, Italy.
| | - Pasquale Capone
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Via Fontana Candida 1, 00078 Monteporzio Catone, Rome, Italy.
| | - Damiano Carbonari
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Via Fontana Candida 1, 00078 Monteporzio Catone, Rome, Italy.
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Via Fontana Candida 1, 00078 Monteporzio Catone, Rome, Italy.
| |
Collapse
|
33
|
Muddathir ARM, Abdallah EI, Khabour OF, Abdelgader RE, Elgari MM. Age- and gender-independent association of glutathione S-transferase null polymorphisms with chronic myeloid leukemia. Bosn J Basic Med Sci 2019; 19:350-354. [PMID: 30995900 DOI: 10.17305/bjbms.2019.4176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023] Open
Abstract
The glutathione S-transferase (GST) genes encode enzymes that mediate the detoxification of xenobiotics by catalyzing the conjugation of glutathione (GSH) to xenobiotic substrates. The aim of the current study is to investigate the association between GSTT1 and GSTM1 polymorphisms and chronic myeloid leukemia (CML) among Sudanese patients. Patients with CML (n = 115) were recruited to the study from the Radiation and Isotope Centre Khartoum (RICK)-Sudan. Healthy individuals (n = 104) were included as controls. Genotyping of GSTT1 and GSTM1 polymorphisms was performed using multiplex PCR. Null deletions in the GSTT1 and GSTM1 genes are common in the Sudanese population (control group), with frequencies of 33.9% and 38.2%, respectively. The frequencies of GSTT1 (OR: 3.25, 95% CI: 1.87-5.65, p < 0.001) and GSTM1 (OR: 2.14, 95% CI: 1.25-3.67, p < 0.005) null genotypes were significantly higher in CML patients vs. controls. The distribution of GSTT1 and GSTM1 null polymorphisms was not different between male and female (p > 0.01) and young and old CML patients (p > 0.05). Hematological parameters were not affected by null polymorphisms in the patient group (p > 0.05). In addition, the frequency of GSTM1 null polymorphism was lower in advanced-phase CML patients compared to chronic-phase patients (p < 0.05). The GSTT1 and GSTM1 null polymorphisms are associated with CML among Sudanese patients, independently of their age and gender.
Collapse
Affiliation(s)
- Abdel Rahim Mahmoud Muddathir
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia; Department of Hematology and Blood Transfusion, Faculty of Medical Laboratory Sciences, Alzaeim Alazhari University, Khartoum, Sudan.
| | | | | | | | | |
Collapse
|
34
|
Huang H, Wu Y, Fu W, Wang X, Zhou L, Xu X, Huang F, Wu Y. Downregulation of Keap1 contributes to poor prognosis and Axitinib resistance of renal cell carcinoma via upregulation of Nrf2 expression. Int J Mol Med 2019; 43:2044-2054. [PMID: 30896860 PMCID: PMC6443340 DOI: 10.3892/ijmm.2019.4134] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling has a protective effect on normal cells. A number of previous studies demonstrated that Keap1/Nrf2 signaling is associated with drug resistance in numerous tumors. The aim of the present study was to investigate the roles of Keap1 in renal cell carcinoma (RCC) and its effect on sensitivity to chemotherapy. Reverse transcription-quantitative polymerase chain reaction was used to detect the mRNA expression of Keap1 in 45 cases of RCC tumors and adjacent normal tissues. A total of five randomly selected patients with RCC, five RCC cell lines and normal renal tubular cells were examined to detect the protein and mRNA expressions of Keap1. The 5-year survival rate was analyzed by Kaplan-Meier analysis. The cell viability was assessed by a Cell Counting kit-8 assay. The cell apoptosis and reactive oxygen species (ROS) were determined by flow cytometry. The expressions of associated proteins were determined by western blot analysis. It was identified that in RCC tissues and RCC cell lines, the expression of Keap1 was downregulated, which was considered to be associated with poor prognosis. In total, 1 µM Axitinib significantly decreased cell viability, promoted ROS release and induced cell apoptosis in ACHN cells. Silencing Keap1 was able to reverse the inhibitory effect of Axitinib and enhance the protein expressions of Nrf2, NAD(P)H dehydrogenase [quinone] 1 and heme oxygenase 1. However, silencing Nrf2 increased the cell sensitivity to Axitinib. Under Axitinib condition, overexpressing Nrf2 was able to increase cell viability; however, overexpressing Keap1 resulted in an opposite effect. Keap1 serves as a tumor suppressor; its low expression was associated with poor prognosis and a decreased sensitivity of RCC cells to Axitinib. A possible mechanism underlying Axitinib resistance may involve Nrf2 overexpression.
Collapse
Affiliation(s)
- Haipeng Huang
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Yunhong Wu
- Medical Records Information Management Division, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Weijin Fu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoming Wang
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Liquan Zhou
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Xiaolong Xu
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Fu Huang
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Yi Wu
- Department of Dermatology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| |
Collapse
|
35
|
Kural C, Kaya Kocdogan A, Şimşek GG, Oğuztüzün S, Kaygın P, Yılmaz I, Bayram T, Izci Y. Glutathione S-Transferases and Cytochrome P450 Enzyme Expression in Patients with Intracranial Tumors: Preliminary Report of 55 Patients. Med Princ Pract 2019; 28:56-62. [PMID: 30321868 PMCID: PMC6558316 DOI: 10.1159/000494496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 10/15/2018] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Intracranial tumors are one of the most frightening and difficult-to-treat tumor types. In addition to surgery, protocols such as chemotherapy and radiotherapy also take place in the treatment. Glutathione S-transferase (GST) and cytochrome P450 (CYP) enzymes are prominent drug-metabolizing enzymes in the human body. The aim of this study is to show the expression of GSTP1, GSTM1, CYP1A1, and CYP1B1 in different types of brain tumors and compare our results with those in the literature. SUBJECTS AND METHODS The expression of GSTP1, GSTM1, CYP1A1, and CYP1B1 was analyzed using immunostaining in 55 patients with intracranial tumors in 2016-2017. For GST and CYP expression in normal brain tissue, samples of a portion of surrounding normal brain tissue as well as a matched far neighbor of tumor tissue were used. The demographic features of the patients were documented and the expression results compared. RESULTS The mean age of the patients was 46.72 years; 29 patients were female and 26 were male. Fifty-seven specimens were obtained from 55 patients. Among them, meningioma was diagnosed in 12, metastases in 12, glioblastoma in 9, and pituitary adenoma in 5. The highest GSTP1, GSTM1, and CYP-1A1 expressions were observed in pituitary adenomas. The lowest GSTP1 expression was detected in glioblastomas and the lowest CYP1B1 expression in pituitary adenomas. CONCLUSION GSTP1 and CYP expression is increased in intracranial tumors. These results should be confirmed with a larger series and different enzyme subtypes.
Collapse
Affiliation(s)
- Cahit Kural
- Department of Neurosurgery, University of Health Sciences and Gulhane Education and Research Hospital, Ankara, Turkey
| | | | - Gulcin Güler Şimşek
- Department of Pathology, University of Health Sciences and Kecioren Education and Research Hospital, Ankara, Turkey
| | | | - Pınar Kaygın
- Department of Biology, Kırıkkale University, Kırıkkale, Turkey
| | - Irmak Yılmaz
- Department of Biology, Kırıkkale University, Kırıkkale, Turkey
| | - Tugbag Bayram
- Department of Biology, Kırıkkale University, Kırıkkale, Turkey
| | - Yusuf Izci
- Department of Neurosurgery, University of Health Sciences and Gulhane Education and Research Hospital, Ankara, Turkey,
| |
Collapse
|
36
|
Radic TM, Coric VM, Pljesa-Ercegovac MS, Basta-Jovanovic GM, Radojevic-Skodric SM, Dragicevic DP, Matic MG, Bogdanovic LM, Dzamic ZM, Simic TP, Savic-Radojevic AR. Concomitance of Polymorphisms in Glutathione Transferase Omega Genes Is Associated with Risk of Clear Cell Renal Cell Carcinoma. TOHOKU J EXP MED 2018; 246:35-44. [PMID: 30224590 DOI: 10.1620/tjem.246.35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glutathione S-transferases (GSTs), a superfamily of multifunctional enzymes, play an important role in the onset and progression of renal cell carcinoma (RCC). However, novel GST omega class (GSTO), consisting of GSTO1-1 and GSTO2-2 isoenzymes, has not been studied in RCC yet. Two coding single nucleotide polymorphisms (SNPs) supposedly affect their functions: GSTO1*C419A (rs4925) causing alanine to aspartate substitution (*A140D) and GSTO2*A424G (rs156697) causing asparagine to aspartate substitution (*N142D), and have been associated with several neurodegenerative diseases and cancers. Functional relevance of yet another GSTO2 polymorphism, identified at the 5' untranslated (5'UTR) gene region (GSTO2*A183G, rs2297235), has not been clearly discerned so far. Therefore, we aimed to assess the effect of specific GSTO1 and GSTO2 gene variants, independently and in interaction with established risk factors (smoking, obesity and hypertension) on the risk for the most aggressive RCC subtype, the clear cell RCC (ccRCC). Genotyping was performed in 239 ccRCC patients and 350 matched controls, while plasma levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a biomarker of oxidative DNA damage, were determined by ELISA. As a result, combined effect of all three variant genotypes exhibited almost 3-fold risk of RCC development. Additionally, this association was confirmed at the haplotype level [variant GSTO1*A/GSTO2*G (rs156697)/GSTO2*G (rs2297235) haplotype], suggesting a potential role of those variants in propensity to RCC. Regarding the gene-environment interactions, variant GSTO2*G (rs156697) homozygous smokers are at higher ccRCC risk. Association in terms of oxidative DNA damage was found for GSTO2 polymorphism in 5'UTR and 8-OHdG. In conclusion, the concomitance of GSTO polymorphisms may influence ccRCC risk.
Collapse
Affiliation(s)
- Tanja M Radic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Vesna M Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Marija S Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Gordana M Basta-Jovanovic
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Sanja M Radojevic-Skodric
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Dejan P Dragicevic
- Faculty of Medicine, University of Belgrade.,Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade
| | - Marija G Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Ljiljana M Bogdanovic
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Zoran M Dzamic
- Faculty of Medicine, University of Belgrade.,Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade
| | - Tatjana P Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Ana R Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| |
Collapse
|
37
|
Prejanò M, Marino T, Russo N. On the Inhibition Mechanism of Glutathione Transferase P1 by Piperlongumine. Insight From Theory. Front Chem 2018; 6:606. [PMID: 30619815 PMCID: PMC6296316 DOI: 10.3389/fchem.2018.00606] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
Piperlongumine (PL) is an anticancer compound whose activity is related to the inhibition of human glutathione transferase of pi class (GSTP1) overexpressed in cancerous tumors and implicated in the metabolism of electrophilic compounds. In the present work, the inhibition mechanism of hydrolyzed piperlongumine (hPL) has been investigated employing QM and QM/MM levels of theory. The potential energy surfaces (PESs) underline the contributions of Tyr residue close to G site in the catalytic pocket of the enzyme. The proposed mechanism occurs through a one-step process represented by the nucleophilic addition of the glutathione thiol to electrophilic species giving rise to the simultaneous C-S and H-C bonds formation. Both the used methods give barrier heights (19.8 and 21.5 kcal mol−1 at QM/MM and QM, respectively) close to that experimentally measured for the C-S bond formations (23.8 kcal mol−1).
Collapse
Affiliation(s)
- Mario Prejanò
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, Italy
| | - Tiziana Marino
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, Italy
| | - Nino Russo
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
38
|
Glutathione Transferases: Potential Targets to Overcome Chemoresistance in Solid Tumors. Int J Mol Sci 2018; 19:ijms19123785. [PMID: 30487385 PMCID: PMC6321424 DOI: 10.3390/ijms19123785] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 12/14/2022] Open
Abstract
Multifunctional enzymes glutathione transferases (GSTs) are involved in the development of chemoresistance, thus representing a promising target for a novel approach in cancer treatment. This superfamily of polymorphic enzymes exhibits extraordinary substrate promiscuity responsible for detoxification of numerous conventional chemotherapeutics, at the same time regulating signaling pathways involved in cell proliferation and apoptosis. In addition to upregulated GST expression, different cancer cell types have a unique GST signature, enabling targeted selectivity for isoenzyme specific inhibitors and pro-drugs. As a result of extensive research, certain GST inhibitors are already tested in clinical trials. Catalytic properties of GST isoenzymes are also exploited in bio-activation of specific pro-drugs, enabling their targeted accumulation in cancer cells with upregulated expression of the appropriate GST isoenzyme. Moreover, the latest approach to increase specificity in treatment of solid tumors is development of GST pro-drugs that are derivatives of conventional anti-cancer drugs. A future perspective is based on the design of new drugs, which would selectively target GST overexpressing cancers more prone to developing chemoresistance, while decreasing side effects in off-target cells.
Collapse
|
39
|
Dong SC, Sha HH, Xu XY, Hu TM, Lou R, Li H, Wu JZ, Dan C, Feng J. Glutathione S-transferase π: a potential role in antitumor therapy. Drug Des Devel Ther 2018; 12:3535-3547. [PMID: 30425455 PMCID: PMC6204874 DOI: 10.2147/dddt.s169833] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glutathione S-transferase π (GSTπ) is a Phase II metabolic enzyme that is an important facilitator of cellular detoxification. Traditional dogma asserts that GSTπ functions to catalyze glutathione (GSH)-substrate conjunction to preserve the macromolecule upon exposure to oxidative stress, thus defending cells against various toxic compounds. Over the past 20 years, abnormal GSTπ expression has been linked to the occurrence of tumor resistance to chemotherapy drugs, demonstrating that this enzyme possesses functions beyond metabolism. This revelation reveals exciting possibilities in the realm of drug discovery, as GSTπ inhibitors and its prodrugs offer a feasible strategy in designing anticancer drugs with the primary purpose of reversing tumor resistance. In connection with the authors' current research, we provide a review on the biological function of GSTπ and current developments in GSTπ-targeting drugs, as well as the prospects of future strategies.
Collapse
Affiliation(s)
- Shu-Chen Dong
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huan-Huan Sha
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Xiao-Yue Xu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Tian-Mu Hu
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Rui Lou
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huizi Li
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jian-Zhong Wu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Chen Dan
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jifeng Feng
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| |
Collapse
|
40
|
Bhat MA, Gandhi G. Glutathione S-transferase P1 gene polymorphisms and susceptibility to coronary artery disease in a subgroup of north Indian population. J Genet 2018; 96:927-932. [PMID: 29321351 DOI: 10.1007/s12041-017-0863-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The present study aimed to investigate the association of g.313A>G and g.341C>T polymorphisms of GSTP1 with coronary artery disease (CAD) in a subgroup of north Indian population. In the present case-control study, CAD patients (n = 200) and age-matched, sex-matched and ethnicity-matched healthy controls (n = 200) were genotyped for polymorphisms in GSTP1 using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Genotype distribution of g.313A>Gand g.341C>T polymorphisms of GSTP1 gene was significantly different between cases and controls (P = 0.005 and 0.024, respectively). Binary logistic regression analysis showed significant association of A/G (odds ratio (OR): 1.6, 95% CI: 1.08-2.49, P = 0.020) and G/G (OR: 3.1, 95% CI: 1.41-6.71, P = 0.005) genotypes of GSTP1 g.313A>G, and C/T (OR: 5.8, 95% CI: 1.26-26.34, P = 0.024) genotype of GSTP1 g.341C>T with CAD. The A/G and G/G genotypes of g.313A>G and C/T genotype of g.341C>T conferred 6.5-fold increased risk for CAD (OR: 6.5, 95% CI: 1.37-31.27, P = 0.018).Moreover, the recessive model of GSTP1 g.313A>G is the best fit inheritance model to predict the susceptible gene effect (OR: 2.3, 95% CI: 1.11-4.92, P = 0.020). In conclusion, statistically significant associations of GSTP1 g.313A>G (A/G, G/G) and g.341C>T (C/T) genotypes with CAD were observed.
Collapse
Affiliation(s)
- M A Bhat
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143 005, India.
| | | |
Collapse
|
41
|
Dasari S, Ganjayi MS, Meriga B. Glutathione S-transferase is a good biomarker in acrylamide induced neurotoxicity and genotoxicity. Interdiscip Toxicol 2018; 11:115-121. [PMID: 31719782 PMCID: PMC6829684 DOI: 10.2478/intox-2018-0007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/13/2017] [Indexed: 01/19/2023] Open
Abstract
Glutathione S-transferases (GSTs) are major defence enzymes of the antioxidant enzymatic system. Cytosolic GSTs are more involved in the detoxification than mitochondrial and microsomal GSTs. GSTs are localized in the cerebellum and hippocampus of the rat brain. Acrylamide (AC) is a well assessed neurotoxin of both animals and humans and it produces skeletal muscle weakness and ataxia. AC is extensively used in several industries such as cosmetic, paper, textile, in ore processing, as soil conditioners, flocculants for waste water treatment and it is present in daily consumed food products, like potato chips, French fries, bread, breakfast cereals and beverages like coffee; it is detected on tobacco smoking. GST acts as a biomarker in response to acrylamide. AC can interact with DNA and therefore generate mutations. In rats, low level expression of glutathione S-trasferase (GST) decreases both memory and life span. The major aim of this review is to provide better information on the antioxidant role of GST against AC induced neurotoxicity and genotoxicity.
Collapse
Affiliation(s)
- Sreenivasulu Dasari
- Department of Biochemistry, Sri Venkateswara University, Tirupati-517502, Andhra Pradesh, India
| | - Muni Swamy Ganjayi
- Department of Biochemistry, Sri Venkateswara University, Tirupati-517502, Andhra Pradesh, India
| | - Balaji Meriga
- Department of Biochemistry, Sri Venkateswara University, Tirupati-517502, Andhra Pradesh, India
| |
Collapse
|
42
|
Liu H, Yang Z, Zang L, Wang G, Zhou S, Jin G, Yang Z, Pan X. Downregulation of Glutathione S-transferase A1 suppressed tumor growth and induced cell apoptosis in A549 cell line. Oncol Lett 2018; 16:467-474. [PMID: 29928434 DOI: 10.3892/ol.2018.8608] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/18/2018] [Indexed: 01/24/2023] Open
Abstract
Glutathione S-transferase A1 (GSTA1) is a phase II detoxification enzyme and serves a crucial role in anti-cancer drug resistance. In our previous study, GSTA1 was identified to be highly expressed in various subtypes of non-small-cell lung cancer cell lines compared with human embryonic lung fibroblast cell line MRC-5. The aim of the present study was to investigate the effect of GSTA1 expression on the proliferation and apoptosis of A549 cells. GSTA1 expression was knocked down or with overexpressed using lentivirus particles. Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to assess the protein, and mRNA levels of GSTA1 in A549 cells, respectively. The effect of GSTA1 manipulation on cell proliferation and apoptosis were investigated in vitro using MTT assays, Hoechst 33258 staining and flow cytometry, and in vivo using A549 cell line xenografts in nude mice. The results of the western blot analysis and RT-qPCR revealed that stable cell models of GSTA1 knockdown, and overexpression were established. The data of the MTT assay indicated that the downregulation of GSTA1 significantly inhibited cell proliferation compared with si-control-transfected cells. These si-GSTA1 A549 cells exhibited typical morphological changes of apoptosis, including chromatin condensation and shrunken nuclei compared with the si-control counterparts. An AnnexinV-fluorescein isothiocyanate assay verified that the downregulation of GSTA1 significantly induced cell apoptosis in vitro. In addition, overexpression of GSTA1 significantly promoted tumor growth in vivo. Accordingly, downregulation of GSTA1 suppressed tumor growth. In conclusion, GSTA1 plays an important role in regulation of cell proliferation and cell apoptosis in A549 cell line.
Collapse
Affiliation(s)
- Huan Liu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhouping Yang
- Department of Pharmacy, Guangdong 999 Brain Hospital, Guangzhou, Guangdong 510510, P.R. China
| | - Linquan Zang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Guixiang Wang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Sigui Zhou
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Guifang Jin
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhicheng Yang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Xuediao Pan
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
43
|
Silva M, Carvalho MDG. Detoxification enzymes: cellular metabolism and susceptibility to various diseases. Rev Assoc Med Bras (1992) 2018; 64:307-310. [DOI: 10.1590/1806-9282.64.04.307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/24/2017] [Indexed: 11/22/2022] Open
Affiliation(s)
- Marcelo Silva
- Hospital Universitário Clementino Fraga Filho, Brazil
| | | |
Collapse
|
44
|
Neal M, Richardson JR. Time to get Personal: A Framework for Personalized Targeting of Oxidative Stress in Neurotoxicity and Neurodegenerative Disease. CURRENT OPINION IN TOXICOLOGY 2018; 7:127-132. [PMID: 30272040 DOI: 10.1016/j.cotox.2018.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The annual cost for neurological disorders in the United States was $789 billion in 2014, and with an aging population these numbers are expected to significantly increase in the next 50 years [1]. Neurodegenerative diseases make up a significant portion of these costs. Neurodegenerative diseases are characterized by the loss of neuronal populations in specific regions of the brain. Although the cause is still unknown for most of these diseases, both genetic and environmental factors are thought to play important roles. There are multiple convergent mechanisms underlying the unique susceptibility of neurons to degeneration, including aging, inflammation, mitochondrial dysfunction, and oxidative stress. Oxidative stress (OS) is of particular importance because evidence indicates that the neuronal populations lost in neurodegenerative diseases are particular susceptible to OS. OS is a complex neurotoxic mechanism that arises from excessive generation of free radicals such as reactive oxygen species (ROS), reduction in anti-oxidant factors, or a combination of the two. A complex interplay between the endogenous susceptibility of the brain, genetic factors, and environmental exposures leads to the harmful generation of OS in the brain and contributes significantly to the initiation and/or progression of neurodegeneration. Unfortunately, therapeutics for neurodegenerative diseases have consistently failed in clinical trials. Thus, a better understanding of the interplay between genetic susceptibility and common molecular mechanisms of environmental contributors to OS generation could aid in elucidation of novel therapeutic strategies for neurodegenerative diseases. This review will explore the current picture of oxidative stress in the brain as it relates to neurotoxicity, specifically exploring common mechanisms behind the endogenous susceptibility of the brain to OS, genetic susceptibility and environmental exposures leading to neurotoxicity, to identify precision/personalized medicine approaches for improving therapeutic outcome.
Collapse
Affiliation(s)
- Matthew Neal
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Diseases and Aging, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Jason R Richardson
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Diseases and Aging, Northeast Ohio Medical University, Rootstown, OH 44272
| |
Collapse
|
45
|
Tecza K, Pamula-Pilat J, Lanuszewska J, Butkiewicz D, Grzybowska E. Pharmacogenetics of toxicity of 5-fluorouracil, doxorubicin and cyclophosphamide chemotherapy in breast cancer patients. Oncotarget 2018; 9:9114-9136. [PMID: 29507678 PMCID: PMC5823653 DOI: 10.18632/oncotarget.24148] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/02/2018] [Indexed: 01/28/2023] Open
Abstract
The differences in patients' response to the same medication, toxicity included, are one of the major problems in breast cancer treatment. Chemotherapy toxicity makes a significant clinical problem due to decreased quality of life, prolongation of treatment and reinforcement of negative emotions associated with therapy. In this study we evaluated the genetic and clinical risk factors of FAC chemotherapy-related toxicities in the group of 324 breast cancer patients. Selected genes and their polymorphisms were involved in FAC drugs transport (ABCB1, ABCC2, ABCG2,SLC22A16), metabolism (ALDH3A1, CBR1, CYP1B1, CYP2C19, DPYD, GSTM1, GSTP1, GSTT1, MTHFR,TYMS), DNA damage recognition, repair and cell cycle control (ATM, ERCC1, ERCC2, TP53, XRCC1). The multifactorial risk models that combine genetic risk modifiers and clinical characteristics were constructed for 12 toxic symptoms. The majority of toxicities was dependent on the modifications in components of more than one pathway of FAC drugs, while the impact level of clinical factors was comparable to the genetic ones. For the carriers of multiple high risk factors the chance of developing given symptom was significantly elevated which proved the factor-dosage effect. We found the strongest associations between concurrent presence of clinical factors - overall and recurrent anemia, nephrotoxicity and early nausea and genetic polymorphisms in genes responsible for DNA repair, drugs metabolism and transport pathways. These results indicate the possibility of selection of the patients with expected high tolerance to FAC treatment and consequently with high chance of chemotherapy completion without the dose reduction, treatment delays and decline in the quality of life.
Collapse
Affiliation(s)
- Karolina Tecza
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Jolanta Pamula-Pilat
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Joanna Lanuszewska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Dorota Butkiewicz
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Ewa Grzybowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| |
Collapse
|
46
|
Kiliç M, Ada AO, Oğuztüzün S, Demirağ F, Çelik S, Biçakçioğlu P, Işcan M. Polymorphisms and Protein Expressions of Glutathione S-Transferase M1 and T1 in Non-Small Cell Lung Cancer. Turk J Pharm Sci 2017; 14:237-242. [PMID: 32454619 DOI: 10.4274/tjps.74745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/23/2017] [Indexed: 12/01/2022]
Abstract
Objectives The deletion polymorphisms of glutathione S-transferase (GST) GSTM1 and GSTT1 genes result in the absence of the corresponding protein, which decreases the detoxification of carcinogens. Studies evaluating polymorphisms and protein expressions in the same patients are limited. Therefore, in this study, we aimed to investigate the association between polymorphisms and protein expressions of GSTM1 and GSTT1 in lung tissues of patients with non-small cell lung cancer (NSCLC). Materials and Methods For protein expression and gene deletion studies, tumor and surrounding tumor free (normal) tissue of 33 patients with NSCLC were used. In paraffin-embedded tissues, immunohistochemistry was used to detect protein expressions, and multiplex polymerase chain reaction amplification was used to identify gene deletions. Results GSTM1 and GSTT1 protein expressions were not detected in patients with GSTM1 and GSTT1 gene deletions, whereas protein expressions were detected in lung tissues of all patients carrying GSTM1 and GSTT1 genes. The protein expression level of GSTT1 was 2.0-fold higher in tumors of patients lacking GSTM1 genes than those with GSTM1 genes (p=0.018). Protein expression of GSTM1 was statistically higher in tumor tissues than in normal tissues of patients with GSTM1 genes (p=0.001). Conclusion These results show that a) there is an association between gene deletions and protein expressions of GSTM1 and GSTT1 in patients with NSCLC, b) in the absence of GSTM1 genes, enhancement of expression of GSTT1 in tumors is likely to show that GSTT1 increases its capacity to detoxify the toxic electrophiles in tumors, and c) GSTM1 protein expression is higher in tumors compared with normal lung tissues of patients with NSCLC.
Collapse
Affiliation(s)
- Murat Kiliç
- Ankara University, Vocational School of Health Services, Department of Pharmacy Services, Ankara, Turkey
| | - Ahmet Oğuz Ada
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara, Turkey
| | - Serpil Oğuztüzün
- Kırıkkale University, Faculty of Arts and Sciences, Department of Biology, Kırıkkale, Turkey
| | - Funda Demirağ
- Atatürk Chest Diseases and Thoracic Surgery Training and Research Hospital, Clinic of Pathology, Ankara, Turkey
| | - Sezgin Çelik
- Yıldız Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, İstanbul, Turkey
| | - Pınar Biçakçioğlu
- Atatürk Chest Diseases and Thoracic Surgery Training and Research Hospital, Clinic of Thoracic Surgery, Ankara, Turkey
| | - Mümtaz Işcan
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara, Turkey
| |
Collapse
|
47
|
Surikova EI, Goroshinskaya IA, Frantsiyants EM, Shalashnaja EV, Nerodo GA, Neskubina IV, Kachesova PS, Nemashkalova LA, Chudilova AV. [The activity of redox-regulatory systems in the primary and recurrence tumors in vulvar cancer]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 63:321-326. [PMID: 28862603 DOI: 10.18097/pbmc20176304321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The activities of superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), glutathione transferase (GST), the content of reduced glutathione (GSH) and malondialdehyde (MDA) were investigated in the samples of the tumor, peritumoral zone and healthy tissue, taken at the line of resection, were obtained from 14 patients with primary squamous cell carcinoma of the vulva, and 13 patients with local recurrence appeared in the period from 3 months to 7 years. by conventional spectrophotometric methods. The content of GSH and the activity of SOD, GPx, GR, GST were significantly increased, while MDA was decreased in the tissue of the primary carcinoma of the vulva in compared with the healthy tissue. Differences in the functioning of the investigated system of enzymes in the peritumoral zone were also revealed in the primary and recurrent tumoral process. Similar but much less pronounced changes were also observed in the recurrent tumor. It is suggested that such dynamics of activity of the studied system with the progression of cancer process can be the result of adaptation to changes in the local biochemical status of healthy (nonmalignant) tissue of the organ carrying the tumor and reflect the metabolic features of the recurrent tumor.
Collapse
Affiliation(s)
- E I Surikova
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| | | | | | - E V Shalashnaja
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| | - G A Nerodo
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| | - I V Neskubina
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| | - P S Kachesova
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| | | | - A V Chudilova
- Rostov Research Institute of Oncology, Rostov-on-Don, Russia
| |
Collapse
|
48
|
Mohana K, Achary A. Human cytosolic glutathione-S-transferases: quantitative analysis of expression, comparative analysis of structures and inhibition strategies of isozymes involved in drug resistance. Drug Metab Rev 2017; 49:318-337. [DOI: 10.1080/03602532.2017.1343343] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Krishnamoorthy Mohana
- Department of Biotechnology, Centre for Research, Kamaraj College of Engineering and Technology, Virudhunagar, India
| | - Anant Achary
- Department of Biotechnology, Centre for Research, Kamaraj College of Engineering and Technology, Virudhunagar, India
| |
Collapse
|
49
|
Georgakis ND, Karagiannopoulos DA, Thireou TN, Eliopoulos EE, Labrou NE, Tsoungas PG, Koutsilieris MN, Clonis YD. Concluding the trilogy: The interaction of 2,2'-dihydroxy-benzophenones and their carbonyl N-analogues with human glutathione transferase M1-1 face to face with the P1-1 and A1-1 isoenzymes involved in MDR. Chem Biol Drug Des 2017; 90:900-908. [PMID: 28440951 DOI: 10.1111/cbdd.13011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/27/2017] [Accepted: 04/14/2017] [Indexed: 02/04/2023]
Abstract
A series of 2,2'-dihydroxybenzophenones and their carbonyl N-analogues were studied as potential inhibitors against human glutathione transferase M1-1 (hGSTM1-1) purified from recombinant E. coli. Their screening revealed an inhibition against hGSTM1-1 within a range of 0-42% (25 μM). The IC50 values for the two stronger ones, 16 and 13, were 53.5 ± 5.6 μΜ and 28.5 ± 2.5 μΜ, respectively. The results were compared with earlier ones for isoenzymes hGSTP1-1 and hGSTA1-1 involved in MDR. All but one bind more strongly to A1-1, than M1-1 and P1-1, the latter being a poor binder. An order of potency A1-1 > > M1-1 > P1-1 meritted 13, 14 and 16 as the most potent inhibitors with hGSTM1-1. Enzyme kinetics with hGSTM1-1 (Km(CDNB) 213 ± 10 μΜ and Km(GSH) 303 ± 11 μΜ) revealed a competitive modality for 16 (Ki(16) = 22.3 ± 1.1 μΜ) and a mixed one for 13 versus CDNB (Ki(13) = 33.3 ± 1.6 μM for the free enzyme and Ki(13) ' = 17.7 ± 1.7 μM for the enzyme-CDNB complex). 5- or 5'-Bromo- or phenyl-substituted (but not in combination) inhibitors, having a H-bonded oxime weakly acidic group of a small volume, are optimal candidates for binding hGSTM1-1. The outcome of the isoenzyme trilogy identified good binder leads for the investigated GSTs involved in MDR.
Collapse
Affiliation(s)
- Nikolaos D Georgakis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | | | - Trias N Thireou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Elias E Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Nikolaos E Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Petros G Tsoungas
- Laboratory of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | - Michael N Koutsilieris
- Department of Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Yannis D Clonis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
50
|
Wang HB, Jin XL, Zheng JF, Wang F, Dai F, Zhou B. Developing piperlongumine-directed glutathione S-transferase inhibitors by an electrophilicity-based strategy. Eur J Med Chem 2017; 126:517-525. [PMID: 27914365 DOI: 10.1016/j.ejmech.2016.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/29/2016] [Accepted: 11/14/2016] [Indexed: 01/10/2023]
Abstract
We report a case of successful design of glutathione S-transferase (GST) inhibitors via a natural product-inspired and electrophilicity-based strategy. Based on this strategy, a novel piperlongumine analog (PL-13) bearing a para-trifluoromethyl group and an α-chlorine on its aromatic and lactam rings, respectively, surfaced as a promising GST inhibitor, thereby overcoming cisplatin resistance in lung cancer A549 cells.
Collapse
Affiliation(s)
- Hai-Bo Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Xiao-Ling Jin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Jia-Fang Zheng
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fu Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China.
| |
Collapse
|