1
|
Zhu Y, Chen Y, Wang Y, Zhu Y, Wang H, Zuo M, Wang J, Li Y, Chen X. Glutathione S-transferase-Pi 1 protects cells from irradiation-induced death by inhibiting ferroptosis in pancreatic cancer. FASEB J 2024; 38:e70033. [PMID: 39258853 DOI: 10.1096/fj.202400373rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
Glutathione S-transferase-Pi 1 (GSTP1) is an isozyme that plays a key role in detoxification and antioxidative damage. It also confers resistance to tumor therapy. However, the specific role of GSTP1 in radiotherapy resistance in pancreatic cancer (PC) is not known. In this study, we investigated how GSTP1 imparts radioresistance in PC. The findings of previous studies and this study revealed that ionizing radiation (IR) induces ferroptosis in pancreatic cancer cells, primarily by upregulating the expression of ACSL4. Our results showed that after IR, GSTP1 prolonged the survival of pancreatic cancer cells by inhibiting ferroptosis but did not affect apoptosis. The expression of GSTP1 reduced cellular ferroptosis by decreasing the levels of ACSL4 and increasing the GSH content. These changes increase the resistance of pancreatic cancer cells and xenograft tumors to IR. Our findings indicate that ferroptosis participates in irradiation-induced cell death and that GSTP1 prevents IR-induced death of pancreatic cancer cells by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Yifan Chen
- Department of Nuclear Medicine, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Yuling Wang
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Yuchun Zhu
- Department of Nuclear Medicine, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Hongyan Wang
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Mengzhe Zuo
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Jianliang Wang
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Yonggang Li
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuelian Chen
- Department of Radiology, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| |
Collapse
|
2
|
Vella V, Ditsiou A, Chalari A, Eravci M, Wooller SK, Gagliano T, Bani C, Kerschbamer E, Karakostas C, Xu B, Zhang Y, Pearl FM, Lopez G, Peng L, Stebbing J, Klinakis A, Giamas G. Kinome-Wide Synthetic Lethal Screen Identifies PANK4 as a Modulator of Temozolomide Resistance in Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306027. [PMID: 38353396 PMCID: PMC11022721 DOI: 10.1002/advs.202306027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/23/2023] [Indexed: 02/17/2024]
Abstract
Temozolomide (TMZ) represents the cornerstone of therapy for glioblastoma (GBM). However, acquisition of resistance limits its therapeutic potential. The human kinome is an undisputable source of druggable targets, still, current knowledge remains confined to a limited fraction of it, with a multitude of under-investigated proteins yet to be characterized. Here, following a kinome-wide RNAi screen, pantothenate kinase 4 (PANK4) isuncovered as a modulator of TMZ resistance in GBM. Validation of PANK4 across various TMZ-resistant GBM cell models, patient-derived GBM cell lines, tissue samples, as well as in vivo studies, corroborates the potential translational significance of these findings. Moreover, PANK4 expression is induced during TMZ treatment, and its expression is associated with a worse clinical outcome. Furthermore, a Tandem Mass Tag (TMT)-based quantitative proteomic approach, reveals that PANK4 abrogation leads to a significant downregulation of a host of proteins with central roles in cellular detoxification and cellular response to oxidative stress. More specifically, as cells undergo genotoxic stress during TMZ exposure, PANK4 depletion represents a crucial event that can lead to accumulation of intracellular reactive oxygen species (ROS) and subsequent cell death. Collectively, a previously unreported role for PANK4 in mediating therapeutic resistance to TMZ in GBM is unveiled.
Collapse
Affiliation(s)
- Viviana Vella
- Department of Biochemistry and BiomedicineSchool of Life SciencesUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | - Angeliki Ditsiou
- Department of Biochemistry and BiomedicineSchool of Life SciencesUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | - Anna Chalari
- Center of Basic ResearchBiomedical Research Foundation of the Academy of AthensAthens11527Greece
| | - Murat Eravci
- Department of Biochemistry and BiomedicineSchool of Life SciencesUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | - Sarah K. Wooller
- School of Life SciencesBioinformatics GroupUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | | | - Cecilia Bani
- Department of Biochemistry and BiomedicineSchool of Life SciencesUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | | | - Christos Karakostas
- Center of Basic ResearchBiomedical Research Foundation of the Academy of AthensAthens11527Greece
| | - Bin Xu
- Cancer CenterRenmin Hospital of Wuhan UniversityWuhanHubei430064China
| | - Yongchang Zhang
- Department of Medical OncologyLung Cancer and Gastrointestinal UnitHunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan430064China
| | - Frances M.G. Pearl
- School of Life SciencesBioinformatics GroupUniversity of Sussex, FalmerBrightonBN1 9QGUK
| | - Gianluca Lopez
- Division of PathologyFondazione IRCCS Ca' Granda – Ospedale Maggiore PoliclinicoMilan20122Italy
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilan20122Italy
| | - Ling Peng
- Department of Respiratory DiseaseZhejiang Provincial People's HospitalHangzhouZhejiang310003China
| | - Justin Stebbing
- Department of Life SciencesAnglia Ruskin UniversityEast RoadCambridgeCB1 1PTUK
| | - Apostolos Klinakis
- Center of Basic ResearchBiomedical Research Foundation of the Academy of AthensAthens11527Greece
| | - Georgios Giamas
- Department of Biochemistry and BiomedicineSchool of Life SciencesUniversity of Sussex, FalmerBrightonBN1 9QGUK
| |
Collapse
|
3
|
Cui J, Li L, Wei S, Wei Y, Gong Y, Yan H, Yu Y, Lin X, Qin H, Li G, Yi L. Involvement of GSTP1 in low dose radiation-induced apoptosis in GM12878 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116128. [PMID: 38387144 DOI: 10.1016/j.ecoenv.2024.116128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Low-dose ionizing radiation-induced protection and damage are of great significance among radiation workers. We aimed to study the role of glutathione S-transferase Pi (GSTP1) in low-dose ionizing radiation damage and clarify the impact of ionizing radiation on the biological activities of cells. RESULTS In this study, we collected peripheral blood samples from healthy adults and workers engaged in radiation and radiotherapy and detected the expression of GSTP1 by qPCR. We utilized γ-rays emitted from uranium tailings as a radiation source, with a dose rate of 14 μGy/h. GM12878 cells subjected to this radiation for 7, 14, 21, and 28 days received total doses of 2.4, 4.7, 7.1, and 9.4 mGy, respectively. Subsequent analyses, including flow cytometry, MTS, and other assays, were performed to assess the ionizing radiation's effects on cellular biological functions. In peripheral blood samples collected from healthy adults and radiologic technologist working in a hospital, we observed a decreased expression of GSTP1 mRNA in radiation personnel compared to the healthy controls. In cultured GM12878 cells exposed to low-dose ionizing radiation from uranium tailings, we noted significant changes in cell morphology, suppression of proliferation, delay in cell cycle progression, and increased apoptosis. These effects were partially reversed by overexpression of GSTP1. Moreover, low-dose ionizing radiation increased GSTP1 gene methylation and downregulated GSTP1 expression. Furthermore, low-dose ionizing radiation affected the expression of GSTP1-related signaling molecules. CONCLUSIONS This study shows that low-dose ionizing radiation damages GM12878 cells and affects their proliferation, cell cycle progression, and apoptosis. In addition, GSTP1 plays a modulating role under low-dose ionizing radiation damage conditions. Low-dose ionizing radiation affects the expression of Nrf2, JNK, and other signaling molecules through GSTP1.
Collapse
Affiliation(s)
- Jian Cui
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Linwei Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuanyun Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yaqi Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hongxia Yan
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yueqiu Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guoqing Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital, Institute of Cardiovascular Disease, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
4
|
Cammarota AL, Falco A, Basile A, Molino C, Chetta M, D’Angelo G, Marzullo L, De Marco M, Turco MC, Rosati A. Pancreatic Cancer-Secreted Proteins: Targeting Their Functions in Tumor Microenvironment. Cancers (Basel) 2023; 15:4825. [PMID: 37835519 PMCID: PMC10571538 DOI: 10.3390/cancers15194825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a ravaging disease with a poor prognosis, requiring a more detailed understanding of its biology to foster the development of effective therapies. The unsatisfactory results of treatments targeting cell proliferation and its related mechanisms suggest a shift in focus towards the inflammatory tumor microenvironment (TME). Here, we discuss the role of cancer-secreted proteins in the complex TME tumor-stroma crosstalk, shedding lights on druggable molecular targets for the development of innovative, safer and more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Anna Lisa Cammarota
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Antonia Falco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Anna Basile
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Carlo Molino
- General Surgery Unit, A.O.R.N. Cardarelli, 80131 Naples, Italy;
| | - Massimiliano Chetta
- Medical and Laboratory Genetics Unit, A.O.R.N., Cardarelli, 80131 Naples, Italy;
| | - Gianni D’Angelo
- Department of Computer Science, University of Salerno, 84084 Fisciano, Italy;
| | - Liberato Marzullo
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Margot De Marco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Maria Caterina Turco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Alessandra Rosati
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
5
|
Zaid MA, Dalmizrak O, Teralı K, Ozer N. Mechanistic insights into the inhibition of human placental glutathione S-transferase P1-1 by abscisic and gibberellic acids: An integrated experimental and computational study. J Mol Recognit 2023; 36:e3050. [PMID: 37555623 DOI: 10.1002/jmr.3050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/10/2023] [Accepted: 07/23/2023] [Indexed: 08/10/2023]
Abstract
The interactions of the classic phytohormones gibberellic acid (gibberellin A3 , GA3 ) and abscisic acid (dormin, ABA), which antagonistically regulate several developmental processes and stress responses in higher plants, with human placental glutathione S-transferase P1-1 (hpGSTP1-1), an enzyme that plays a role in endo- or xenobiotic detoxification and regulation of cell survival and apoptosis, were investigated. The inhibitory potencies of ABA and GA3 against hpGSTP1, as well as the types of inhibition and the kinetic parameters, were determined by making use of both enzyme kinetic graphs and SPSS nonlinear regression models. The structural basis for the interaction between hpGSTP1-1 and phytohormones was predicted with the aid of molecular docking simulations. The IC50 values of ABA and GA3 were 5.3 and 5.0 mM, respectively. Both phytohormones inhibited hpGSTP1-1 in competitive manner with respect to the cosubstrates GSH and CDNB. When ABA was the inhibitor at [CDNB]f -[GSH]v and at [GSH]f -[CDNB]v , Vm , Km , and Ki values were statistically estimated to be 205 ± 16 μmol/min-mg protein, 1.32 ± 0.18 mM, 1.95 ± 0.25 mM and 175 ± 6 μmol/min-mg protein, 0.85 ± 0.06 mM, 1.85 ± 0.16 mM, respectively. On the other hand, the kinetic parameters Vm , Km , and Ki obtained with GA3 at [CDNB]f -[GSH]v and at [GSH]f -[CDNB]v were found to be 303 ± 14 μmol/min-mg protein, 1.77 ± 0.13 mM, 3.38 ± 0.26 mM and 249 ± 7 μmol/min-mg protein, 1.43 ± 0.07 mM, 2.89 ± 0.19 mM, respectively. Both phytohormones had the potential to engage in hydrogen-bonding and electrostatic interactions with the key residues that line the G- and H-sites of the enzyme's catalytic center. Inhibitory actions of ABA/GA3 on hpGSTP1-1 may guide medicinal chemists through the structure-based design of novel antineoplastic agents. It should be noted, however, that the same interactions may also render fetuses vulnerable to the potentially toxic effects of xenobiotics and noxious endobiotics.
Collapse
Affiliation(s)
| | - Ozlem Dalmizrak
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Kerem Teralı
- Department of Medical Biochemistry, Faculty of Medicine, Cyprus International University, Nicosia, Cyprus
| | - Nazmi Ozer
- Department of Biochemistry, Faculty of Pharmacy, Girne American University, Kyrenia, Cyprus
| |
Collapse
|
6
|
Azarova I, Polonikov A, Klyosova E. Molecular Genetics of Abnormal Redox Homeostasis in Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054738. [PMID: 36902173 PMCID: PMC10003739 DOI: 10.3390/ijms24054738] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Numerous studies have shown that oxidative stress resulting from an imbalance between the production of free radicals and their neutralization by antioxidant enzymes is one of the major pathological disorders underlying the development and progression of type 2 diabetes (T2D). The present review summarizes the current state of the art advances in understanding the role of abnormal redox homeostasis in the molecular mechanisms of T2D and provides comprehensive information on the characteristics and biological functions of antioxidant and oxidative enzymes, as well as discusses genetic studies conducted so far in order to investigate the contribution of polymorphisms in genes encoding redox state-regulating enzymes to the disease pathogenesis.
Collapse
Affiliation(s)
- Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Alexey Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Correspondence:
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| |
Collapse
|
7
|
Silva MAP, Figueiredo DBS, Lara JR, Paschoalinotte EE, Braz LG, Braz MG. Evaluation of genetic instability, oxidative stress, and metabolism-related gene polymorphisms in workers exposed to waste anesthetic gases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:9609-9623. [PMID: 36057057 DOI: 10.1007/s11356-022-22765-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Professionals who work in operating rooms (ORs) may be exposed daily to waste anesthetic gases (WAGs) due to the use of inhalational anesthetics. Considering the controversial findings related to genetic damage and redox status in addition to a lack of knowledge about the effect of polymorphisms in genes related to phase I and II detoxification upon occupational exposure to WAGs, this cross-sectional study is the first to jointly evaluate biomarkers of genetic instability, oxidative stress, and susceptibility genes in professionals occupationally exposed to high trace amounts of halogenated (≥ 7 ppm) and nitrous oxide (165 ppm) anesthetics in ORs and in individuals not exposed to WAGs (control group). Elevated rates of buccal micronucleus (MN) and nuclear bud (NBUD) were observed in the exposure group and in professionals exposed aged more than 30 years. Exposed males showed a higher antioxidant capacity, as determined by the ferric reducing antioxidant power (FRAP), than exposed females; exposed females had higher frequencies of MN and NBUD than nonexposed females. Genetic instability (MN) was observed in professionals with greater weekly WAG exposure, and those exposed for longer durations (years) exhibited oxidative stress (increased lipid peroxidation and decreased FRAP). Polymorphisms in metabolic genes (cytochrome P450 2E1 (CYP2E1) and glutathione S-transferases (GSTs)) did not exert an effect, except for the effects of the GSTP1 (rs1695) AG/GG polymorphism on FRAP (both groups) and GSTP1 AG/GG and GSTT1 null polymorphisms, which were associated with greater FRAP values in exposed males. Minimizing WAG exposure is necessary to reduce impacts on healthcare workers.
Collapse
Affiliation(s)
- Mariane A P Silva
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil
| | - Drielle B S Figueiredo
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil
| | - Juliana R Lara
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil
| | - Eloisa E Paschoalinotte
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil
| | - Leandro G Braz
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil
| | - Mariana G Braz
- Medical School - São Paulo State University (UNESP), Prof. Mario Rubens G. Montenegro Av. Botucatu, São Paulo, 18618-687, Brazil.
| |
Collapse
|
8
|
Praeruptorin B inhibits osteoclastogenesis by targeting GSTP1 and impacting on the S-glutathionylation of IKKβ. Biomed Pharmacother 2022; 154:113529. [PMID: 36030586 DOI: 10.1016/j.biopha.2022.113529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Osteoporosis a common disease in postmenopausal women which contains significant impact on the living quality of women. With the aging of the population, the number of patients suffer from osteoporosis has shown a significant increase. Given the limitations of clinical drugs for the treatment of osteoporosis, natural extracts with small side effects have a great application prospect in the treatment of osteoporosis. Praeruptorin B (Pra-B), is one of the main components found in the roots of Peucedanum praeruptorum Dunn and exhibits anti-inflammatory effects. However, there is no research on the influence of Pra-B on osteoporosis. Here, we showed that Pra-B can dose-dependently suppress osteoclastogenesis without cytotoxicity. Receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL)-induced the nuclear import of P65 was inhibited by Pra-B, which indicated the suppressive effect of Pra-B on NF-κB signaling. Further, Pra-B enhanced the expression of Glutathione S-transferase Pi 1 (GSTP1) and promoted the S-glutathionylation of IKKβ to inhibit the nuclear translocation of P65. Moreover, in vivo experiments showed that Pra-B considerably attenuated the bone loss in ovariectomy (OVX)-induced mice. Collectively, our studies revealed that Pra-B suppress the NF-κB signaling targeting GSTP1 to rescued RANKL-induced osteoclastogenesis in vitro and OVX-induced bone loss in vivo, supporting the potential of Pra-B for treating osteoporosis in the future.
Collapse
|
9
|
Genomic and functional insights into the diversification of the elongation factor eEF1Bγ in fungi. FUNGAL BIOL REV 2022. [DOI: 10.1016/j.fbr.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
10
|
Zhang Z, Xu L, Huang L, Li T, Wang JY, Ma C, Bian X, Ren X, Li H, Wang X. Glutathione S-Transferase Alpha 4 Promotes Proliferation and Chemoresistance in Colorectal Cancer Cells. Front Oncol 2022; 12:887127. [PMID: 35936694 PMCID: PMC9346510 DOI: 10.3389/fonc.2022.887127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/31/2022] [Indexed: 11/16/2022] Open
Abstract
Glutathione S-transferase alpha 4 (GSTA4) is a phase II detoxifying enzyme that is overexpressed in colorectal cancer (CRC) and regulated by the oncogenic transcription factor AP-1. However, the role of GSTA4 in these CRC cells remains unclear. In this study, we investigated the roles of GSTA4 in the CRC cells by inactivating GSTA4 in HCT116 human CRC cells (Defined as HCT116ΔGSTA4) using the CRISPR/Cas9 gene editing. Cell proliferation, clonogenicity, and susceptibility to chemotherapeutic drugs were analyzed in vitro and in a xenograft model. The results showed that loss of GSTA4 significantly decreased cell proliferation and clonogenicity, whereas it increased intracellular reactive oxygen species and cell susceptibility to 5-fluorouracil (5-FU) and oxaliplatin. Additionally, exposure of HCT116ΔGSTA4 cells to 5-FU increased the expression of γH2AX, a hallmark of double-stranded DNA breaks. In contrast, no remarkably increased γH2AX was noted in oxaliplatin-treated HCT116ΔGSTA4 cells compared with HCT116 cells. Moreover, loss of GSTA4 blocked the AKT and p38 MAPK pathways, leading to proliferative suppression. Finally, the xenograft model showed decreased tumor size for HCT116ΔGSTA4 cells compared with HCT116 cells, confirming in vitro findings. These findings suggest that GSTA4 is capable of promoting proliferation, tumorigenesis, and chemoresistance and is a potential target for CRC therapy.
Collapse
Affiliation(s)
- Zhanhu Zhang
- Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Lili Xu
- Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Lin Huang
- Department of Gastroenterology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Tianqi Li
- Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jane Y. Wang
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Chunhua Ma
- Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Xiaoyun Bian
- Department of Gastroenterology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Xiaoyan Ren
- Department of Pathology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Haibo Li
- Department of Clinical Laboratory, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Xingmin Wang
- Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- *Correspondence: Xingmin Wang,
| |
Collapse
|
11
|
Li X, Kong R, Li Y, Huang J, Zhou X, Li S, Cheng H. Carrier-free nanomedicine for enhanced photodynamic tumor therapy through glutathione S-transferase inhibition. Chem Commun (Camb) 2022; 58:3917-3920. [PMID: 35237781 DOI: 10.1039/d2cc00235c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Antioxidant-defense systems of tumor cells protect them from oxidative damage. Herein, a carrier-free nanomedicine is developed based on chlorine e6 (Ce6) and coniferyl ferulate (Con), which inhibits glutathione S-transferase (GST) activity to hamper antioxidant systems and amplify intracellular oxidative stress for enhanced photodynamic therapy.
Collapse
Affiliation(s)
- Xinyu Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Renjiang Kong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Yanmei Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Jiaqi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Xiang Zhou
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| | - Shiying Li
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China.
| |
Collapse
|
12
|
Lin H, Sun W, Zeng T, Li H, Xu C, Chen Y, Yin W. Identification of fosaprepitant as a novel GSTP1 inhibitor through structure-based virtual screening, molecular dynamics simulation, and biological evaluation. NEW J CHEM 2022. [DOI: 10.1039/d1nj04597k] [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]
Abstract
The workflow of virtual screening for the discovery of GSTP1 inhibitors.
Collapse
Affiliation(s)
- Hao Lin
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Wenxiu Sun
- Department of pharmacy, Lingbi people's Hospital, Suzhou, Anhui, China
| | - Tao Zeng
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Hengda Li
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Chenming Xu
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Yan Chen
- The Affiliated cancer hospital of Nanjing Medical University, Jiangsu cancer hospital, Nanjing, China
| | - Wu Yin
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Rodrigues ACBDC, Bomfim LM, Neves SP, Soares MBP, Dias RB, Valverde LF, Rocha CAG, Costa EV, da Silva FMA, Rocha WC, Koolen HHF, Bezerra DP. Tingenone and 22-hydroxytingenone target oxidative stress through downregulation of thioredoxin, leading to DNA double-strand break and JNK/p38-mediated apoptosis in acute myeloid leukemia HL-60 cells. Biomed Pharmacother 2021; 142:112034. [PMID: 34411914 DOI: 10.1016/j.biopha.2021.112034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/27/2021] [Accepted: 08/07/2021] [Indexed: 11/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most lethal form of leukemia. Standard anti-AML treatment remains almost unchanged for decades. Tingenone (TG) and 22-hydroxytingenone (22-HTG) are quinonemethide triterpenes found in the Amazonian plant Salacia impressifolia (Celastraceae), with cytotoxic properties in different histological types of cancer cells. In the present work, we investigated the anti-AML action mechanism of TG and 22-HTG in the AML HL-60 cell line. Both compounds exhibited potent cytotoxicity in a panel of cancer cell lines. Mechanistic studies found that TG and 22-HTG reduced cell growth and caused the externalization of phosphatidylserine, the fragmentation of internucleosomal DNA and the loss of mitochondrial transmembrane potential in HL-60 cells. In addition, pre-incubation with Z-VAD(OMe)-FMK, a pan-caspase inhibitor, prevented TG- and 22-HTG-induced apoptosis, indicating cell death by apoptosis via a caspase-dependent pathway. The analysis of the RNA transcripts of several genes indicated the interruption of the cellular antioxidant system, including the downregulation of thioredoxin, as a target for TG and 22-HTG. The application of N-acetyl-cysteine, an antioxidant, completely prevented apoptosis induced by TG and 22-HTG, indicating activation of the apoptosis pathway mediated by oxidative stress. Moreover, TG and 22-HTG induced DNA double-strand break and phosphorylation of JNK2 (T183/Y185) and p38α (T180/Y182), and co-incubation with SP 600125 (JNK/SAPK inhibitor) and PD 169316 (p38 MAPK inhibitor) partially prevented apoptosis induced by TG and 22-HTG. Together, these data indicate that TG and 22-HTG are new candidate for anti-AML therapy targeting thioredoxin.
Collapse
Affiliation(s)
| | - Larissa M Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Sara P Neves
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil; SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, BA 41650-010, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil; Department of Clinical Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, Bahia 40301-155, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil; Department of Clinical Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, Bahia 40301-155, Brazil
| | - Emmanoel V Costa
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas 69080-900, Brazil
| | - Felipe M A da Silva
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas 69080-900, Brazil
| | - Waldireny C Rocha
- Health and Biotechnology Institute, Federal University of Amazonas (UFAM), Coari, Amazonas 69460-000, Brazil
| | - Hector H F Koolen
- Metabolomics and Mass Spectrometry Research Group, Amazonas State University (UEA), Manaus, Amazonas 690065-130, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil.
| |
Collapse
|
14
|
Ściskalska M, Milnerowicz H. Activity of glutathione S-transferase and its π isoenzyme in the context of single nucleotide polymorphism in the GSTP1 gene (rs1695) and tobacco smoke exposure in the patients with acute pancreatitis and healthy subjects. Biomed Pharmacother 2021; 140:111589. [PMID: 34130200 DOI: 10.1016/j.biopha.2021.111589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress associated with the course of acute pancreatitis (AP) can cause changes in the involvement of antioxidants, which can result in the increased production of free radicals with pro-inflammatory potential. Through its noncatalytic activity, the glutathione S-transferase and its π isoenzyme (GST-π), apart from cellular xenobiotics detoxification, are involved in the regulation of cellular signalling, metabolism and apoptosis. This study aimed to evaluate the impact of SNP rs1695 in the GSTP1 gene on GST and GST-π activity in healthy subjects and patients with acute pancreatitis (AP). The concentration of glutathione (GSH) as an important component of the antioxidant system, necessary for environmental xenobiotics detoxification by GST, and malonyldialdehyde (MDA) as a marker of oxidative stress induced by inflammation were also assessed. SNP was examined in 39 AP patients and 51 healthy subjects using PCR-RFLP methods. GST activity (in plasma and erythrocyte lysate) and GST-π activity (in erythrocyte lysate) were measured using the spectrophotometric method with 1-chloro-2,4-dinitrobenzene and ethacrynic acid as substrate, respectively. Blood GSH concentration was measured using the Patterson method. Concentrations of high-sensitive C-reactive protein (hs-CRP) and MDA were measured using commercial tests. In the blood of non-smoking AP patients with GG genotypes for SNP rs1695 in the GSTP1 gene, the lowest GST-π activity was shown. It was accompanied by the lowest hsCRP concentration in this group. In the blood of smoking healthy subjects with AG genotype, a decrease in GST-π activity was noted compared to non-smokers from this group. However, in the blood of smokers with AP, a gradually decreasing GST-π activity was noted in individuals with AA genotype, which was associated with the increasing MDA concentration. It confirms the role of GST-π in the neutralization of oxidative stress induced by the exposure to smoke xenobiotics.
Collapse
Affiliation(s)
- Milena Ściskalska
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland.
| | - Halina Milnerowicz
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland.
| |
Collapse
|
15
|
Wang Q, Bin C, Xue Q, Gao Q, Huang A, Wang K, Tang N. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis 2021; 12:426. [PMID: 33931597 PMCID: PMC8087704 DOI: 10.1038/s41419-021-03718-4] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Increasing evidence supports that ferroptosis plays an important role in tumor growth inhibition. Sorafenib, originally identified as an inhibitor of multiple oncogenic kinases, has been shown to induce ferroptosis in hepatocellular carcinoma (HCC). However, some hepatoma cell lines are less sensitive to sorafenib-induced ferroptotic cell death. Glutathione S-transferase zeta 1 (GSTZ1), an enzyme in the catabolism of phenylalanine, suppresses the expression of the master regulator of cellular redox homeostasis nuclear factor erythroid 2-related factor 2 (NRF2). This study aimed to investigate the role and underlying molecular mechanisms of GSTZ1 in sorafenib-induced ferroptosis in HCC. GSTZ1 was significantly downregulated in sorafenib-resistant hepatoma cells. Mechanistically, GSTZ1 depletion enhanced the activation of the NRF2 pathway and increased the glutathione peroxidase 4 (GPX4) level, thereby suppressing sorafenib-induced ferroptosis. The combination of sorafenib and RSL3, a GPX4 inhibitor, significantly inhibited GSTZ1-deficient cell viability and promoted ferroptosis and increased ectopic iron and lipid peroxides. In vivo, the combination of sorafenib and RSL3 had a synergic therapeutic effect on HCC progression in Gstz1-/- mice. In conclusion, this finding demonstrates that GSTZ1 enhanced sorafenib-induced ferroptosis by inhibiting the NRF2/GPX4 axis in HCC cells. Combination therapy of sorafenib and GPX4 inhibitor RSL3 may be a promising strategy in HCC treatment.
Collapse
Affiliation(s)
- Qiujie Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Cheng Bin
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qiang Xue
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingzhu Gao
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021; 10:antiox10050701. [PMID: 33946704 PMCID: PMC8146591 DOI: 10.3390/antiox10050701] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
In humans, the glutathione S-transferases (GST) protein family is composed of seven members that present remarkable structural similarity and some degree of overlapping functionalities. GST proteins are crucial antioxidant enzymes that regulate stress-induced signaling pathways. Interestingly, overactive GST proteins are a frequent feature of many human cancers. Recent evidence has revealed that the biology of most GST proteins is complex and multifaceted and that these proteins actively participate in tumorigenic processes such as cell survival, cell proliferation, and drug resistance. Structural and pharmacological studies have identified various GST inhibitors, and these molecules have progressed to clinical trials for the treatment of cancer and other diseases. In this review, we discuss recent findings in GST protein biology and their roles in cancer development, their contribution in chemoresistance, and the development of GST inhibitors for cancer treatment.
Collapse
|