1
|
Appell ML, Hindorf U, Almer S, Haglund S. Response to azathioprine treatment in autoimmune hepatitis is dependent on glutathione transferase genotypes. Dig Liver Dis 2025; 57:885-892. [PMID: 39863504 DOI: 10.1016/j.dld.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Azathioprine (AZA) is part of the standard treatment for autoimmune hepatitis (AIH). The first step in the complex bioconversion of AZA to active metabolites is mediated by glutathione transferases (GSTs). AIMS Elucidate the association between GSTM1 and GSTT1 copy number variation (CNV), genetic variation in GSTA2, GSTP1, and inosine-triphosphate-pyrophosphatase, and the response to AZA in AIH. METHODS Genotyping was performed in AIH patients (n = 131) on AZA, and in a Swedish background population (n = 283). Thiopurine metabolites in blood erythrocytes were determined by high performance liquid chromatography. RESULTS GSTM1 and GSTT1 CNV were associated with treatment response to AZA. Gene deletion of GSTM1-but not of GSTT1-was associated with the liver transaminase levels. None of the studied genetic variants were associated with the thiopurine metabolite concentrations, suggesting non-enzymatic mechanisms of GSTM1 and GSTT1 in the context of AZA efficacy in AIH. The prevalence of GSTM1 and GSTT1 CNV genotypes was similar in AIH and in the background population. CONCLUSION This study shows the effects of GSTM1 and GSTT1 CNV on AZA efficacy in AIH, not previously described. It also elaborates on the impact of the definition of treatment response, on the importance of the various GSTs studied. Furthermore, the GSTM1 and GSTT1 CNV frequencies previously reported in European populations were confirmed.
Collapse
Affiliation(s)
- Malin Lindqvist Appell
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Ulf Hindorf
- Department of Gastroenterology and Nutrition, University Hospital Lund, Lund, Sweden.
| | - Sven Almer
- Centre for Digestive Health, Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Sofie Haglund
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Laboratory Medicine, Region Jönköping County, Jönköping, Sweden.
| |
Collapse
|
2
|
Timme K, Inyang I, White HE, Keating AF. Diet-induced obesity alters the ovarian chemical biotransformation and oxidative stress response proteins both basally and in response to 7,12-dimethylbenz[a]anthracene exposure. Toxicol Sci 2025; 204:9-19. [PMID: 39910959 PMCID: PMC11879017 DOI: 10.1093/toxsci/kfae150] [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] [Indexed: 02/07/2025] Open
Abstract
7,12-Dimethylbenz[a]anthracene (DMBA) is a polycyclic aromatic hydrocarbon that causes female infertility via DNA damage, and the ovary has the capacity to mitigate DMBA exposure via the action of proteins including the glutathione S-transferase (GST) family. Due to previous findings of DNA damage and a reduced ovarian chemical biotransformation response to DMBA exposure in hyperphagia-induced obese mice, this study investigated the hypothesis that diet-induced obesity would hamper the ovarian biotransformative response to DMBA exposure. Six-week-old C57BL6/J mice were fed either a normal rodent diet (L) or a high fat high sucrose diet (O) until the O group was ∼30% heavier than the L. Both L and O mice were exposed to either corn oil (C) or DMBA (1 mg/kg) for 7 d. Liver weight was increased (P < 0.05) in obese mice exposed to DMBA but no effect on spleen weight, uterine weight, ovary weight, estrous cyclicity, or circulating 17β-estradiol and progesterone were observed. Primordial and preantral follicle numbers were higher (P < 0.05) in the obese mice and there was a tendency (P = 0.055) for higher antral follicles in DMBA-exposed obese mice. The ovarian proteome was identified by LC-MS/MS analysis to be altered both by diet-induced obesity and by DMBA exposure with changes observed in levels of proteins involved in oocyte development and chemical biotransformation, including GST isoform pi. Fewer proteins were affected by the combined exposure of diet and DMBA than by a single treatment, indicating that physiological status impacts the response to DMBA exposure.
Collapse
Affiliation(s)
- Kelsey Timme
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States
| | - Imaobong Inyang
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States
| | - Hunter E White
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States
| |
Collapse
|
3
|
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
|
4
|
Timme K, González-Alvarez ME, Keating AF. Pre-pubertal obesity compromises ovarian oxidative stress, DNA repair and chemical biotransformation. Toxicol Appl Pharmacol 2024; 489:116981. [PMID: 38838792 DOI: 10.1016/j.taap.2024.116981] [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: 03/11/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Obesity in adult females impairs fertility by altering oxidative stress, DNA repair and chemical biotransformation. Whether prepubertal obesity results in similar ovarian impacts is under-explored. The objective of this study was to induce obesity in prepubertal female mice and assess puberty onset, follicle number, and abundance of oxidative stress, DNA repair and chemical biotransformation proteins basally and in response to 7,12-dimethylbenz(a)anthracene (DMBA) exposure. DMBA is a polycyclic aromatic hydrocarbon that has been shown to be ovotoxic. Lactating dams (C57BL6J) were fed either a normal rodent containing 3.5% kCal from fat (lean), or a high fat diet comprised of 60% kCal from fat, and 9% kCal from sucrose. The offspring were weaned onto the diet of their dam and exposed at postnatal day 35 to either corn oil or DMBA (1 mg/kg) for 7 d via intraperitoneal injection. Mice on the HFD had reduced (P < 0.05) age at puberty onset as measured by vaginal opening but DMBA did not impact puberty onset. Heart, spleen, kidney, uterus and ovary weight were increased (P < 0.05) by obesity and liver weight was increased (P < 0.05) by DMBA exposure in obese mice. Follicle number was largely unaffected by obesity or DMBA exposure, with the exception of primary follicle number, which were higher (P < 0.05) in lean DMBA exposed and obese control relative to lean control mice. There were also greater numbers (P < 0.05) of corpora lutea in obese relative to lean mice. In lean mice, DMBA exposure reduced (P < 0.05) the level of CYP2E1, EPHX1, GSTP1, BRCA1, and CAT but this DMBA-induced reduction was absent in obese mice. Basally, obesity reduced (P < 0.05) the abundance of CYP2E1, EPHX1, GSTP1, BRCA1, SOD1 and CAT. There was greater (P < 0.05) fibrotic staining in obese DMBA-exposed ovaries and PPP2CA was decreased (P < 0.05) in growing follicles by both obesity and DMBA exposure. Thus, prepubertal obesity alters the capacity of the ovary to respond to DNA damage, ovotoxicant exposure and oxidative stress.
Collapse
Affiliation(s)
- Kelsey Timme
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA, USA.
| |
Collapse
|
5
|
Baliova M, Jursky F. Manganese- and zinc-coordinated interaction of Schistosoma japonicum glutathione S-transferase with neurotransmitter transporters GlyT1 and GAT3 in vitro. Exp Parasitol 2024; 259:108721. [PMID: 38369179 DOI: 10.1016/j.exppara.2024.108721] [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: 08/17/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Glutathione S-transferases (GSTs) are a family of multifunctional isoenzymes involved in the neutralization of toxic compounds, drug resistance and several other cellular functions. The glutathione S-transferase enzyme of Schistosoma japonicum (SjGST-26) plays a role in human schistosomiasis and is also a frequently used fusion partner in mammalian and bacterial expression and pull-down systems. GSTs seem not to be naturally associated with metal ions. Exceptionally, in vitro, metal binding sites have been previously described in some schistosome GSTs; however, their possible physiological role is unclear. Molecules of several neurotransmitter transporters also contain a regulatory zinc binding site, which affects their transport cycle. Here we show that among several metals, manganese and zinc are able to induce a specific protein interaction of SjGST-26 with the glycine transporter GlyT1 and the GABA transporter GAT3 in vitro. The results suggest that metal-binding sites on SjGST-26 and neurotransmitter transporters might function in metal-coordinated interactions with other metalloproteins. Our results additionally indicate that the presence of metal ions in SjGST-26-based GST protein pull-down assays may lead to a false-positive interaction if the potential interacting target is the metalloprotein.
Collapse
Affiliation(s)
- Martina Baliova
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Dubravska Cesta 21, 845 51, Bratislava, Slovakia
| | - Frantisek Jursky
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Dubravska Cesta 21, 845 51, Bratislava, Slovakia.
| |
Collapse
|
6
|
Xu T, Pan Y, Ding Q, Cao F, Chang K, Qiu J, Zhuge H, Hao L, Wei H, Si C, Dou X, Li S. The micro-743a-3p-GSTM1 pathway is an endogenous protective mechanism against alcohol-related liver disease in mice. Cell Mol Biol Lett 2024; 29:35. [PMID: 38475733 DOI: 10.1186/s11658-024-00557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND AND AIMS Epidemiological evidence suggests that the phenotype of glutathione S-transferase mu 1 (GSTM1), a hepatic high-expressed phase II detoxification enzyme, is closely associated with the incidence of alcohol-related liver disease (ALD). However, whether and how hepatic GSTM1 determines the development of ALD is largely unclear. This study was designed to elucidate the role and potential mechanism(s) of hepatic GSTM1 in the pathological process of ALD. METHODS GSTM1 was detected in the liver of various ALD mice models and cultured hepatocytes. Liver-specific GSTM1 or/and micro (miR)-743a-3p deficiency mice were generated by adenoassociated virus-8 delivered shRNA, respectively. The potential signal pathways involving in alcohol-regulated GSTM1 and GSTM1-associated ALD were explored via both genetic manipulation and pharmacological approaches. RESULTS GSTM1 was significantly upregulated in both chronic alcohol-induced mice liver and ethanol-exposed murine primary hepatocytes. Alcohol-reduced miR-743a-3p directly contributed to the upregulation of GSTM1, since liver specific silencing miR-743a-3p enhanced GSTM1 and miR-743a-3p loss protected alcohol-induced liver dysfunctions, which was significantly blocked by GSTM1 knockdown. GSTM1 loss robustly aggravated alcohol-induced hepatic steatosis, oxidative stress, inflammation, and early fibrotic-like changes, which was associated with the activation of apoptosis signal-regulating kinase 1 (ASK1), c-Jun N-terminal kinase (JNK), and p38. GSTM1 antagonized ASK1 phosphorylation and its downstream JNK/p38 signaling pathway upon chronic alcohol consumption via binding with ASK1. ASK1 blockage significantly rescued hepatic GSTM1 loss-enhanced disorders in alcohol-fed mice liver. CONCLUSIONS Chronic alcohol consumption-induced upregulation of GSTM1 in the liver provides a feedback protection against hepatic steatosis and liver injury by counteracting ASK1 activation. Down-regulation of miR-743a-3p improves alcohol intake-induced hepatic steatosis and liver injury via direct targeting on GSTM1. The miR-743a-3p-GSTM1 axis functions as an innate protective pathway to defend the early stage of ALD.
Collapse
Affiliation(s)
- Tiantian Xu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Yan Pan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Feiwei Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Hui Zhuge
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Liuyi Hao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Haibin Wei
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Caijuan Si
- Department of Clinical Nutrition, School of Medicine, Affiliated Zhejiang Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China.
- Department of Clinical Nutrition, School of Medicine, Affiliated Zhejiang Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
7
|
Zhou N, Zhang Y, Jiao Y, Nan J, Xia A, Mu B, Lin G, Li X, Zhang S, Yang S, Li L. Discovery of a novel pyroptosis inhibitor acting though modulating glutathionylation to suppress NLRP3-related signal pathway. Int Immunopharmacol 2024; 127:111314. [PMID: 38081102 DOI: 10.1016/j.intimp.2023.111314] [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: 09/23/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
Pyroptosis is a proinflammatory type of regulated cell death and has been involved in many pathological processes. Inhibition of pyroptosis is thought to be a promising strategy for the treatment of related diseases. Here, we performed a phenotypic screening against NLRP3-dependent pyroptosis and obtained the novel compound N77 after structure optimization. N77 showed a half-maximal effective concentration (EC50) of 0.070 ± 0.008 μM against cell pyroptosis induced by nigericin, and exhibited a remarkable ability to prevent NLRP3-dependent inflammasome activation and the release of IL-1β. Chemical proteomics revealed the biological target of N77 to be glutathione-S-transferase Mu 1 (GSTM1); our mechanism of action studies indicated that GSTM1 might act as a negative regulator of NLRP3 inflammasome activation by modulating the glutathionylation of caspase-1. In vivo, N77 substantially alleviated the inflammatory reaction in a pyroptosis-related acute keratitis model. Overall, we identified a novel pyroptosis inhibitor and revealed a new regulatory mechanism of pyroptosis. Our findings suggest an alternative potential therapeutic strategy for pyroptosis-related diseases.
Collapse
Affiliation(s)
- Nenghua Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yun Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Macular Disease Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Jiao
- Laboratory of Anaesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu China
| | - Jinshan Nan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anjie Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Macular Disease Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Mu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Basic Medical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Guifeng Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xun Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Macular Disease Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shanshan Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Lv N, Huang C, Huang H, Dong Z, Chen X, Lu C, Zhang Y. Overexpression of Glutathione S-Transferases in Human Diseases: Drug Targets and Therapeutic Implications. Antioxidants (Basel) 2023; 12:1970. [PMID: 38001822 PMCID: PMC10668987 DOI: 10.3390/antiox12111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione S-transferases (GSTs) are a major class of phase II metabolic enzymes. Besides their essential role in detoxification, GSTs also exert diverse biological activities in the occurrence and development of various diseases. In the past few decades, much research interest has been paid to exploring the mechanisms of GST overexpression in tumor drug resistance. Correspondingly, many GST inhibitors have been developed and applied, solely or in combination with chemotherapeutic drugs, for the treatment of multi-drug resistant tumors. Moreover, novel roles of GSTs in other diseases, such as pulmonary fibrosis and neurodegenerative diseases, have been recognized in recent years, although the exact regulatory mechanisms remain to be elucidated. This review, firstly summarizes the roles of GSTs and their overexpression in the above-mentioned diseases with emphasis on the modulation of cell signaling pathways and protein functions. Secondly, specific GST inhibitors currently in pre-clinical development and in clinical stages are inventoried. Lastly, applications of GST inhibitors in targeting cell signaling pathways and intracellular biological processes are discussed, and the potential for disease treatment is prospected. Taken together, this review is expected to provide new insights into the interconnection between GST overexpression and human diseases, which may assist future drug discovery targeting GSTs.
Collapse
Affiliation(s)
- Ning Lv
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chunyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Haoyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Zhiqiang Dong
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
| | - Xijing Chen
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chengcan Lu
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
- Jiangning Clinical Medical College, Jiangsu University, Nanjing 211100, China
| | - Yongjie Zhang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| |
Collapse
|
9
|
Park SY, Kim KS, Lee WY, Kim CE, Lee S. Integrative Approach to Identifying System-Level Mechanisms of Chung-Sang-Bo-Ha-Hwan's Influence on Respiratory Tract Diseases: A Network Pharmacological Analysis with Experimental Validation. PLANTS (BASEL, SWITZERLAND) 2023; 12:3024. [PMID: 37687271 PMCID: PMC10489874 DOI: 10.3390/plants12173024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
Chung-Sang-Bo-Ha-Hwan (CSBHH) is an herbal prescription widely used to treat various chronic respiratory diseases. To investigate the system-level treatment mechanisms of CSBHH in respiratory tract diseases, we identified 56 active ingredients of CSBHH and evaluated the degree of overlap between their targets and respiratory tract disease-associated proteins. We then investigated the respiratory tract disease-related signaling pathways associated with CSBHH targets. Enrichment analysis showed that the CSBHH targets were significantly associated with various signaling pathways related to inflammation, alveolar structure, and tissue fibrosis. Experimental validation was conducted using phorbol-12-myristate-13-acetate (PMA)-stimulated NCI-H292 cells by analyzing the mRNA expression levels of biomarkers (IL-1β and TNF-α for inflammation; GSTP1, GSTM1, and PTEN for apoptosis) derived from network pharmacological analysis, in addition to the mucin genes MUC5AC and MUC2, to investigate the phlegm-expelling effect of CSBHH. The mRNA expression levels of these genes were consistent with network pharmacological predictions in a concentration-dependent manner. These results suggest that the therapeutic mechanisms of CSBHH in respiratory tract diseases could be attributed to the simultaneous action of multiple active ingredients in the herbal prescription.
Collapse
Affiliation(s)
- Sa-Yoon Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Kang-Sub Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea;
| | - Won-Yung Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
10
|
Chen Q, Guo J, Qiu T, Zhou J. Mechanism of ASK1 involvement in liver diseases and related potential therapeutic targets: A critical pathway molecule worth investigating. J Gastroenterol Hepatol 2023; 38:378-385. [PMID: 36533997 DOI: 10.1111/jgh.16087] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/25/2022] [Accepted: 12/10/2022] [Indexed: 12/30/2022]
Abstract
Since the discovery of apoptosis signal-regulated kinase 1 (ASK1), the signal transduction mechanism and pathophysiological process involved in its regulation have been continuously revealed. Many previous studies have identified that ASK1 is involved and plays a critical role in the development of diseases affecting the nervous, cardiac, renal, and other systems. As a mitogen-activated protein kinase (MAPK) kinase kinase, ASK1 mediates apoptosis, necrosis, inflammation, and other pathological processes by activating its downstream c-Jun N-terminal kinase (JNK)/p38 MAPK. Owing to the important role of ASK1, an increasing number of studies in recent years have focused on its status in liver-related diseases. In this paper, we review the mechanisms and targets of ASK1 in liver-related diseases to emphasize its important role in the development of liver disease.
Collapse
Affiliation(s)
- Qi Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
11
|
Russell TM, Richardson DR. The good Samaritan glutathione-S-transferase P1: An evolving relationship in nitric oxide metabolism mediated by the direct interactions between multiple effector molecules. Redox Biol 2023; 59:102568. [PMID: 36563536 PMCID: PMC9800640 DOI: 10.1016/j.redox.2022.102568] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Glutathione-S-transferases (GSTs) are phase II detoxification isozymes that conjugate glutathione (GSH) to xenobiotics and also suppress redox stress. It was suggested that GSTs have evolved not to enhance their GSH affinity, but to better interact with and metabolize cytotoxic nitric oxide (NO). The interactions between NO and GSTs involve their ability to bind and store NO as dinitrosyl-dithiol iron complexes (DNICs) within cells. Additionally, the association of GSTP1 with inducible nitric oxide synthase (iNOS) results in its inhibition. The function of NO in vasodilation together with studies associating GSTM1 or GSTT1 null genotypes with preeclampsia, additionally suggests an intriguing connection between NO and GSTs. Furthermore, suppression of c-Jun N-terminal kinase (JNK) activity occurs upon increased levels of GSTP1 or NO that decreases transcription of JNK target genes such as c-Jun and c-Fos, which inhibit apoptosis. This latter effect is mediated by the direct association of GSTs with MAPK proteins. GSTP1 can also inhibit nuclear factor kappa B (NF-κB) signaling through its interactions with IKKβ and Iκα, resulting in decreased iNOS expression and the stimulation of apoptosis. It can be suggested that the inhibitory activity of GSTP1 within the JNK and NF-κB pathways may be involved in crosstalk between survival and apoptosis pathways and modulating NO-mediated ROS generation. These studies highlight an innovative role of GSTs in NO metabolism through their interaction with multiple effector proteins, with GSTP1 functioning as a "good Samaritan" within each pathway to promote favorable cellular conditions and NO levels.
Collapse
Affiliation(s)
- Tiffany M Russell
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Australia.
| |
Collapse
|
12
|
Izquierdo AG, Carreira MC, Rodriguez-Carnero G, Perez-Lois R, Seoane LM, Casanueva FF, Crujeiras AB. Gender Dimorphism in Hepatic Carcinogenesis-Related Gene Expression Associated with Obesity as a Low-Grade Chronic Inflammatory Disease. Int J Mol Sci 2022; 23:ijms232315002. [PMID: 36499327 PMCID: PMC9739425 DOI: 10.3390/ijms232315002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) show clear evidence of sexual dimorphism, with a significantly higher incidence in males. Among the determining factors that could explain this sex-based difference, the specific distribution of fat by sex has been suggested as a primary candidate, since obesity is a relevant risk factor. In this context, obesity, considered a low-grade chronic inflammatory pathology and responsible for the promotion of liver disease, could lead to sexual dimorphism in the expression profile of genes related to tumor development. When we compared the expression levels of genes associated with the early stages of carcinogenesis in the liver between male and female diet-induced obesity (DIO) rats, we observed that the expression pattern was similar in obese male and female animals. Interestingly, the SURVIVIN/BIRC5 oncogene showed a higher expression in male DIO rats than in female DIO and lean rats. This trend related to sexual dimorphism was observed in leukocytes from patients with obesity, although the difference was not statistically significant. In conclusion, this study evidenced a similar pattern in the expression of most carcinogenesis-related genes in the liver, except SUVIVIN/BIRC5, which could be a predictive biomarker of liver carcinogenesis predisposition in male patients with obesity.
Collapse
Affiliation(s)
- Andrea G. Izquierdo
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Marcos C. Carreira
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), 28029 Madrid, Spain
- Molecular Endocrinology Group, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Gemma Rodriguez-Carnero
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Raquel Perez-Lois
- Endocrine Physiopathology Group, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Luisa M. Seoane
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), 28029 Madrid, Spain
- Endocrine Physiopathology Group, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Felipe F. Casanueva
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), 28029 Madrid, Spain
- Molecular Endocrinology Group, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Ana B. Crujeiras
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), 28029 Madrid, Spain
- Correspondence: or ; Tel.: +34-981-955-710
| |
Collapse
|
13
|
Raad S, David A, Sagniez M, Paré B, Orfi Z, Dumont NA, Smith MA, Faure C. iPSCs derived from esophageal atresia patients reveal SOX2 dysregulation at the anterior foregut stage. Dis Model Mech 2022; 15:dmm049541. [PMID: 36317486 PMCID: PMC10655818 DOI: 10.1242/dmm.049541] [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: 03/03/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2023] Open
Abstract
A series of well-regulated cellular and molecular events result in the compartmentalization of the anterior foregut into the esophagus and trachea. Disruption of the compartmentalization process leads to esophageal atresia/tracheoesophageal fistula (EA/TEF). The cause of EA/TEF remains largely unknown. Therefore, to mimic the early development of the esophagus and trachea, we differentiated induced pluripotent stem cells (iPSCs) from EA/TEF patients, and iPSCs and embryonic stem cells from healthy individuals into mature three-dimensional esophageal organoids. CXCR4, SOX17 and GATA4 expression was similar in both patient-derived and healthy endodermal cells. The expression of the key transcription factor SOX2 was significantly lower in the patient-derived anterior foregut. We also observed an abnormal expression of NKX2.1 (or NKX2-1) in the patient-derived mature esophageal organoids. At the anterior foregut stage, RNA sequencing revealed the critical genes GSTM1 and RAB37 to be significantly lower in the patient-derived anterior foregut. We therefore hypothesize that a transient dysregulation of SOX2 and the abnormal expression of NKX2.1 in patient-derived cells could be responsible for the abnormal foregut compartmentalization.
Collapse
Affiliation(s)
- Suleen Raad
- Esophageal Development and Engineering Laboratory, CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
| | - Anu David
- Esophageal Development and Engineering Laboratory, CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
| | - Melanie Sagniez
- CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, Quebec H3T 1J4, Canada
| | - Bastien Paré
- CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, Quebec H3T 1J4, Canada
| | - Zakaria Orfi
- CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
| | - Nicolas A. Dumont
- CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
- School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Martin A. Smith
- CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, Quebec H3T 1J4, Canada
| | - Christophe Faure
- Esophageal Development and Engineering Laboratory, CHU Sainte-Justine Research Center, 3175 Côte Sainte-Catherine, Montréal, Quebec H3T 1C5, Canada
- Esophageal Atresia Clinic and Division of Pediatric Gastroenterology Hepatology and Nutrition, CHU Sainte-Justine, 3715 Côte Sainte-Catherine, Université de Montréal, Montréal, Quebec H3T1C5, Canada
| |
Collapse
|
14
|
Levy R, Le TH. Role of GSTM1 in Hypertension, CKD, and Related Diseases across the Life Span. KIDNEY360 2022; 3:2153-2163. [PMID: 36591365 PMCID: PMC9802555 DOI: 10.34067/kid.0004552022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022]
Abstract
Over 20 years after the introduction of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, CKD remains a major public health burden with limited therapeutic options to halt or slow kidney disease progression at all ages. The consensus is that oxidative stress contributes to CKD development and progression. Yet, to date, there is no clear evidence that broad use of antioxidant therapy provides a beneficial effect in CKD. Understanding the specific pathophysiologic mechanisms in those who are genetically most susceptible to oxidative stress is a crucial step to inform therapy in an individualized medicine approach, considering differing exposures and risks across the life span. Glutathione-S-transferase μ 1 (GSTM1) is a phase 2 enzyme involved in inactivation of reactive oxygen species and metabolism of xenobiotics. In particular, those with the highly prevalent GSTM1 null genotype (GSTM1[0/0]) may be more susceptible to kidney disease progression, due to impaired capacity to handle the increased oxidative stress burden in disease states, and might specifically benefit from therapy that targets the redox imbalance mediated by loss of the GSTM1 enzyme. In this review, we will discuss the studies implicating the role of GSTM1 deficiency in kidney and related diseases from experimental rodent models to humans, from the prenatal period through senescence, and the potential underlying mechanism.
Collapse
Affiliation(s)
- Rebecca Levy
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Thu H. Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
15
|
Garriga F, Llavanera M, Viñolas-Vergés E, Recuero S, Tamargo C, Delgado-Bermúdez A, Yeste M. Glutathione S-transferase Mu 3 is associated to in vivo fertility, but not sperm quality, in bovine. Animal 2022; 16:100609. [PMID: 35963103 DOI: 10.1016/j.animal.2022.100609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/01/2022] Open
Abstract
In the dairy breeding industry, pregnancy of dairy cows is essential to initiate milk production, so that high fertility rates are required to increase their productivity. In this regard, sperm proteins that are indicative of sperm quality and/or fertility have become an important target of study. Glutathione S-transferase Mu 3 (GSTM3) has been established as a fertility and sperm quality parameter in humans and pigs and, consequently, it might be a potential biomarker in cattle. For this reason, the present work aimed to determine if GSTM3 could predict sperm quality and in vivo fertility in this species. Sperm quality was assessed with flow cytometry and computer-assisted sperm analysis. Immunoblotting and immunofluorescence analysis were performed to determine the presence and localisation pattern of sperm GSTM3. This enzyme was found to be present in bovine sperm and to be localised along the sperm tail and the equatorial segment of the head. No significant associations between sperm GSTM3 and sperm quality parameters were observed, except a negative association with morphologically abnormal sperm having a coiled tail. In addition, and more relevant, higher levels of GSTM3 in sperm were seen in bulls showing lower in vivo fertility rates. In conclusion, our data evidenced the presence of GSTM3 in bovine sperm. Moreover, we suggest that, despite not being associated with sperm quality, GSTM3 might be an in vivo subfertility biomarker in cattle sperm, and that high levels of this protein could be an indicative of defective spermatogenesis and/or epididymal maturation.
Collapse
Affiliation(s)
- Ferran Garriga
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Marc Llavanera
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Estel Viñolas-Vergés
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Sandra Recuero
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Carolina Tamargo
- Department of Animal Selection and Reproduction, The Regional Agri-Food Research and Development Service of Asturias (SERIDA), ES-33394 Gijón, Spain
| | - Ariadna Delgado-Bermúdez
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), ES-08010 Barcelona, Spain.
| |
Collapse
|
16
|
Jin Y, Tan Y, Zhao P, Guo Y, Chen S, Wu J, Ren Z. Glutathione S-transferase Mu 2 inhibits hepatic steatosis via ASK1 suppression. Commun Biol 2022; 5:326. [PMID: 35388144 PMCID: PMC8986781 DOI: 10.1038/s42003-022-03251-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 03/11/2022] [Indexed: 12/31/2022] Open
Abstract
Hepatic steatosis is the main characteristic of some liver metabolism diseases. However, unclear molecular mechanism of hepatic steatosis impedes the therapy of this hepatic steatosis. Glutathione-S-transferase mu 2 (GSTM2), as a member of phase II drug metabolizing enzymes (DMEs), regulates cellular antioxidant and detoxificant. GSTM2 was highly up-regulated in hepatic steatosis tissues and high-fat diet (HFD) fed mice. Loss-of-function GSTM2 mouse model demonstrated that GSTM2 protected mice from excess fat accumulation. Mechanistically, GSTM2 interacted with ASK1 and suppressed its phosphorylation and the activation of subsequent downstream p38-JNK signalling. Moreover, GSTM2 overexpression in the liver effectively ameliorated hepatic lipid accumulation. Therefore, we identified GSTM2 as an important negative regulator in progression of hepatic steatosis via both its detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling. This study showed potential therapeutic function of the DME in progression of hepatic steatosis. Jin et al. investigate the underlying mechanisms of hepatic steatosis and show that Glutathione-S-transferase mu 2 (GSTM2), which is a drug metabolizing enzyme (DME), is upregulated in hepatic steatosis tissue from mice fed with a high fat diet. They show that GSTM2 is a negative regulator of hepatic steatosis via both detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling, which sheds light on its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China
| | - Yanjie Tan
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Pengxiang Zhao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Yu Guo
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Shilin Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China. .,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China.
| |
Collapse
|
17
|
Gardiner LJ, Carrieri AP, Bingham K, Macluskie G, Bunton D, McNeil M, Pyzer-Knapp EO. Combining explainable machine learning, demographic and multi-omic data to inform precision medicine strategies for inflammatory bowel disease. PLoS One 2022; 17:e0263248. [PMID: 35196350 PMCID: PMC8865677 DOI: 10.1371/journal.pone.0263248] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/15/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis and Crohn’s disease, affect several million individuals worldwide. These diseases are heterogeneous at the clinical, immunological and genetic levels and result from complex host and environmental interactions. Investigating drug efficacy for IBD can improve our understanding of why treatment response can vary between patients. We propose an explainable machine learning (ML) approach that combines bioinformatics and domain insight, to integrate multi-modal data and predict inter-patient variation in drug response. Using explanation of our models, we interpret the ML models’ predictions to infer unique combinations of important features associated with pharmacological responses obtained during preclinical testing of drug candidates in ex vivo patient-derived fresh tissues. Our inferred multi-modal features that are predictive of drug efficacy include multi-omic data (genomic and transcriptomic), demographic, medicinal and pharmacological data. Our aim is to understand variation in patient responses before a drug candidate moves forward to clinical trials. As a pharmacological measure of drug efficacy, we measured the reduction in the release of the inflammatory cytokine TNFα from the fresh IBD tissues in the presence/absence of test drugs. We initially explored the effects of a mitogen-activated protein kinase (MAPK) inhibitor; however, we later showed our approach can be applied to other targets, test drugs or mechanisms of interest. Our best model predicted TNFα levels from demographic, medicinal and genomic features with an error of only 4.98% on unseen patients. We incorporated transcriptomic data to validate insights from genomic features. Our results showed variations in drug effectiveness (measured by ex vivo assays) between patients that differed in gender, age or condition and linked new genetic polymorphisms to patient response variation to the anti-inflammatory treatment BIRB796 (Doramapimod). Our approach models IBD drug response while also identifying its most predictive features as part of a transparent ML precision medicine strategy.
Collapse
Affiliation(s)
- Laura-Jayne Gardiner
- IBM Research Europe—Daresbury, The Hartree Centre, Warrington, United Kingdom
- * E-mail: (APC); (LJG)
| | - Anna Paola Carrieri
- IBM Research Europe—Daresbury, The Hartree Centre, Warrington, United Kingdom
- * E-mail: (APC); (LJG)
| | - Karen Bingham
- REPROCELL Europe Ltd, Glasgow, Scotland, United Kingdom
| | | | - David Bunton
- REPROCELL Europe Ltd, Glasgow, Scotland, United Kingdom
| | - Marian McNeil
- Precision Medicine Scotland Innovation Centre, Teaching and Learning Building, Queen Elizabeth University Hospital, Glasgow, Scotland, United Kingdom
| | | |
Collapse
|
18
|
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
|
19
|
Rodrigues de Souza I, Savio de Araujo-Souza P, Morais Leme D. Genetic variants affecting chemical mediated skin immunotoxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2022; 25:43-95. [PMID: 34979876 DOI: 10.1080/10937404.2021.2013372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The skin is an immune-competent organ and this function may be impaired by exposure to chemicals, which may ultimately result in immune-mediated dermal disorders. Interindividual variability to chemical-induced skin immune reactions is associated with intrinsic individual characteristics and their genomes. In the last 30-40 years, several genes influencing susceptibility to skin immune reactions were identified. The aim of this review is to provide information regarding common genetic variations affecting skin immunotoxicity. The polymorphisms selected for this review are related to xenobiotic-metabolizing enzymes (CYPA1 and CYPB1 genes), antioxidant defense (GSTM1, GSTT1, and GSTP1 genes), aryl hydrocarbon receptor signaling pathway (AHR and ARNT genes), skin barrier function transepidermal water loss (FLG, CASP14, and SPINK5 genes), inflammation (TNF, IL10, IL6, IL18, IL31, and TSLP genes), major histocompatibility complex (MHC) and neuroendocrine system peptides (CALCA, TRPV1, ACE genes). These genes present variants associated with skin immune responses and diseases, as well as variants associated with protecting skin immune homeostasis following chemical exposure. The molecular and association studies focusing on these genetic variants may elucidate their functional consequences and contribution in the susceptibility to skin immunotoxicity. Providing information on how genetic variations affect the skin immune system may reduce uncertainties in estimating chemical hazards/risks for human health in the future.
Collapse
Affiliation(s)
| | | | - Daniela Morais Leme
- Graduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba, Brazil
- National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Institute of Chemistry, Araraquara, Brazil
| |
Collapse
|
20
|
Lan T, Hu Y, Hu F, Li H, Chen Y, Zhang J, Yu Y, Jiang S, Weng Q, Tian S, Ma T, Yang G, Luo D, Wang L, Li K, Piao S, Rong X, Guo J. Hepatocyte glutathione S-transferase mu 2 prevents non-alcoholic steatohepatitis by suppressing ASK1 signaling. J Hepatol 2022; 76:407-419. [PMID: 34656650 DOI: 10.1016/j.jhep.2021.09.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. The advanced stage of NAFLD, non-alcoholic steatohepatitis (NASH), has been recognized as a leading cause of end-stage liver injury for which there are no FDA-approved therapeutic options. Glutathione S-transferase Mu 2 (GSTM2) is a phase II detoxification enzyme. However, the roles of GSTM2 in NASH have not been elucidated. METHODS Multiple RNA-seq analyses were used to identify hepatic GSTM2 expression in NASH. In vitro and in vivo gain- or loss-of-function approaches were used to investigate the role and molecular mechanism of GSTM2 in NASH. RESULTS We identified GSTM2 as a sensitive responder and effective suppressor of NASH progression. GSTM2 was significantly downregulated during NASH progression. Hepatocyte GSTM2 deficiency markedly aggravated insulin resistance, hepatic steatosis, inflammation and fibrosis induced by a high-fat diet and a high-fat/high-cholesterol diet. Mechanistically, GSTM2 sustained MAPK pathway signaling by directly interacting with apoptosis signal-regulating kinase 1 (ASK1). GSTM2 directly bound to the N-terminal region of ASK1 and inhibited ASK1 N-terminal dimerization to subsequently repress ASK1 phosphorylation and the activation of its downstream JNK/p38 signaling pathway under conditions of metabolic dysfunction. CONCLUSIONS These data demonstrated that hepatocyte GSTM2 is an endogenous suppressor that protects against NASH progression by blocking ASK1 N-terminal dimerization and phosphorylation. Activating GSTM2 holds promise as a therapeutic strategy for NASH. CLINICAL TRIAL NUMBER IIT-2021-277. LAY SUMMARY New therapeutic strategies for non-alcoholic steatohepatitis are urgently needed. We identified that the protein GSTM2 exerts a protective effect in response to metabolic stress. Therapies that aim to increase the activity of GSTM2 could hold promise for the treatment of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Tian Lan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yufeng Hu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fengjiao Hu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haonan Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yinghua Chen
- Organ Transplant, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Zhang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Yang Yu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Shuo Jiang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Qiqing Weng
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Song Tian
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tengfei Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guizhi Yang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Duosheng Luo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Lexun Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Kunping Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Shenghua Piao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Xianglu Rong
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Jiao Guo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
21
|
Teslenko I, Watson CJW, Xia Z, Chen G, Lazarus P. Characterization of Cytosolic Glutathione S-Transferases Involved in the Metabolism of the Aromatase Inhibitor, Exemestane. Drug Metab Dispos 2021; 49:1047-1055. [PMID: 34593616 PMCID: PMC11025106 DOI: 10.1124/dmd.121.000635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Exemestane (EXE) is a hormonal therapy used to treat estrogen receptor-positive breast cancer by inhibiting the final step of estrogen biosynthesis catalyzed by the enzyme aromatase. Cysteine conjugates of EXE and its active metabolite 17β-dihydro-EXE (DHE) are the major metabolites found in both the urine and plasma of patients taking EXE. The initial step in cysteine conjugate formation is glutathione conjugation catalyzed by the glutathione S-transferase (GST) family of enzymes. The goal of the present study was to identify cytosolic hepatic GSTs active in the GST-mediated metabolism of EXE and 17β-DHE. Twelve recombinant cytosolic hepatic GSTs were screened for their activity against EXE and 17β-DHE, and glutathionylated EXE and 17β-DHE conjugates were detected by ultra-performance liquid chromatography tandem mass spectrometry. GST α (GSTA) isoform 1, GST μ (GSTM) isoform 3 and isoform 1 were active against EXE, whereas only GSTA1 exhibited activity against 17β-DHE. GSTM1 exhibited the highest affinity against EXE with a Michaelis-Menten constant (KM) value that was 3.8- and 7.1-fold lower than that observed for GSTA1 and GSTM3, respectively. Of the three GSTs, GSTM3 exhibited the highest intrinsic clearance against EXE (intrinsic clearance = 0.14 nl·min-1·mg-1). The KM values observed for human liver cytosol against EXE (46 μM) and 17β-DHE (77 μM) were similar to those observed for recombinant GSTA1 (53 and 30 μM, respectively). Western blot analysis revealed that GSTA1 and GSTM1 composed 4.3% and 0.57%, respectively, of total protein in human liver cytosol; GSTM3 was not detected. These data suggest that GSTA1 is the major hepatic cytosolic enzyme involved in the clearance of EXE and its major active metabolite, 17β-DHE. SIGNIFICANCE STATEMENT: Most previous studies related to the metabolism of the aromatase inhibitor exemestane (EXE) have focused mainly on phase I metabolic pathways and the glucuronidation phase II metabolic pathway. However, recent studies have indicated that glutathionylation is the major metabolic pathway for EXE. The present study is the first to characterize hepatic glutathione S-transferase (GST) activity against EXE and 17β-dihydro-EXE and to identify GST α 1 and GST μ 1 as the major cytosolic GSTs involved in the hepatic metabolism of EXE.
Collapse
Affiliation(s)
- Irina Teslenko
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Christy J W Watson
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Zuping Xia
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Gang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
22
|
Genome-wide identification and expression profiling of glutathione S-transferase family under hypoxia stress in silver sillago (Sillago sihama). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100920. [PMID: 34628157 DOI: 10.1016/j.cbd.2021.100920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 11/22/2022]
Abstract
Glutathione S-transferase (GST) is an important detoxification enzyme in organisms. GSTs play an important role in responding to environmental stresses. This study aimed to identify the GST gene superfamily in silver sillago (Sillago sihama) and analyze its expression pattern under hypoxia stress. A total of 17 GST genes were identified in silver sillago. Phylogenetic analysis showed that the GST gene family contained two subgroups (cytosolic and MAPEGs), and lacked three subgroups (i.e. Pi, Kappa, and MGST2). Phylogenetic and syntenic analysis revealed that GST genes were conserved in evolution. Eight SsGSTs were significantly differentially expressed under hypoxia stress in silver sillago by RNA-seq and qRT-PCR analysis. The expression levels of SsMGST3b, SsGSTO1, SsGSTT1b and SsGSTR2 genes were significantly up-regulated after 4 h of reoxygenation in the gill tissue. In the heart tissue, the expression of SsGSTR3 was significantly up-regulated after 1 h of hypoxia while the expression levels of SsGSTT1b and SsFLAP genes were significantly down-regulated after 4 h of hypoxia. In summary, this study provides for the first time a comprehensive analysis of the GST gene superfamily of silver sillago.
Collapse
|
23
|
Le TH. GSTM1 Gene, Diet, and Kidney Disease: Implication for Precision Medicine?: Recent Advances in Hypertension. Hypertension 2021; 78:936-945. [PMID: 34455814 DOI: 10.1161/hypertensionaha.121.16510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the United States, the prevalence of chronic kidney disease in adults is ≈14%. The mainstay of therapy for chronic kidney disease is angiotensin-converting enzyme inhibitors or angiotensin receptor blockers, but many patients with chronic kidney disease still progress to end-stage kidney disease. Increased oxidative stress is a major molecular underpinning of chronic kidney disease progression. In humans, a common deletion variant of the glutathione-S-transferase μ-1 (GSTM1) gene, the GSTM1 null allele (GSTM1(0)), results in decreased GSTM1 enzymatic activity and is associated with higher levels of oxidative stress. GSTM1 belongs to the superfamily of GSTs that are phase II antioxidant enzymes and are regulated by Nrf2 (nuclear factor erythroid 2-related factor 2). Cruciferous vegetables in general, and broccoli in particular, are rich in glucoraphanin, a precursor of sulforaphane that has been shown to have protective effects against oxidative damage through the activation of Nrf2. This review will highlight recent human and animal studies implicating the role of GSTM1 deficiency in hypertension and kidney disease, and its impact on the effects of cruciferous vegetables on kidney injury and disease progression, illustrating the significance of gene and environment interaction and a potential for targeted precision medicine in the treatment of kidney disease.
Collapse
Affiliation(s)
- Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, NY
| |
Collapse
|
24
|
Redox and Inflammatory Signaling, the Unfolded Protein Response, and the Pathogenesis of Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:333-373. [PMID: 34019276 DOI: 10.1007/978-3-030-68748-9_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Protein folding overload and oxidative stress disrupt endoplasmic reticulum (ER) homeostasis, generating reactive oxygen species (ROS) and activating the unfolded protein response (UPR). The altered ER redox state induces further ROS production through UPR signaling that balances the cell fates of survival and apoptosis, contributing to pulmonary microvascular inflammation and dysfunction and driving the development of pulmonary hypertension (PH). UPR-induced ROS production through ER calcium release along with NADPH oxidase activity results in endothelial injury and smooth muscle cell (SMC) proliferation. ROS and calcium signaling also promote endothelial nitric oxide (NO) synthase (eNOS) uncoupling, decreasing NO production and increasing vascular resistance through persistent vasoconstriction and SMC proliferation. C/EBP-homologous protein further inhibits eNOS, interfering with endothelial function. UPR-induced NF-κB activity regulates inflammatory processes in lung tissue and contributes to pulmonary vascular remodeling. Conversely, UPR-activated nuclear factor erythroid 2-related factor 2-mediated antioxidant signaling through heme oxygenase 1 attenuates inflammatory cytokine levels and protects against vascular SMC proliferation. A mutation in the bone morphogenic protein type 2 receptor (BMPR2) gene causes misfolded BMPR2 protein accumulation in the ER, implicating the UPR in familial pulmonary arterial hypertension pathogenesis. Altogether, there is substantial evidence that redox and inflammatory signaling associated with UPR activation is critical in PH pathogenesis.
Collapse
|
25
|
Decreased Expression of the Host Long-Noncoding RNA-GM Facilitates Viral Escape by Inhibiting the Kinase activity TBK1 via S-glutathionylation. Immunity 2021; 53:1168-1181.e7. [PMID: 33326766 DOI: 10.1016/j.immuni.2020.11.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 07/29/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022]
Abstract
Viruses have evolved multiple strategies to evade elimination by the immune system. Here we examined the contribution of host long noncoding RNAs (lncRNAs) in viral immune evasion. By functional screening of lncRNAs whose expression decreased upon viral infection of macrophages, we identified a lncRNA (lncRNA-GM, Gene Symbol: AK189470.1) that promoted type I interferon (IFN-I) production and inhibited viral replication. Deficiency of lncRNA-GM in mice increased susceptibility to viral infection and impaired IFN-I production. Mechanistically, lncRNA-GM bound to glutathione S-transferase M1 (GSTM1) and blocked GSTM1 interaction with the kinase TBK1, reducing GSTM1-mediated S-glutathionylation of TBK1. Decreased S-glutathionylation enhanced TBK1 activity and downstream production of antiviral mediators. Viral infection reprogrammed intracellular glutathione metabolism and furthermore, an oxidized glutathione mimetic could inhibit TBK1 activity and promote viral replication. Our findings reveal regulation of TBK1 by S-glutathionylation and provide insight into the viral mediated metabolic changes that impact innate immunity and viral evasion.
Collapse
|
26
|
Li X, Lü Z, Wang C, Li K, Xu F, Xu P, Niu Y. Induction of Apoptosis in Cancer Cells by Glutathione Transferase Inhibitor Mediated Hydrophobic Tagging Molecules. ACS Med Chem Lett 2021; 12:720-725. [PMID: 34055217 DOI: 10.1021/acsmedchemlett.0c00627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/29/2021] [Indexed: 11/30/2022] Open
Abstract
The abnormally high expression of glutathione transferases is closely associated with cancer incidence and drug resistance. By introducing a hydrophobic moiety to the inhibitor structure, we organized a series of degraders of glutathione transferases and demonstrated them potently inducing apoptosis in cancer cells, presenting their pharmacological potential in cancer therapy.
Collapse
Affiliation(s)
- Xiaona Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Zirui Lü
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Cong Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Kebin Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Fengrong Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Ping Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Yan Niu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
27
|
van de Wetering C, Elko E, Berg M, Schiffers CHJ, Stylianidis V, van den Berge M, Nawijn MC, Wouters EFM, Janssen-Heininger YMW, Reynaert NL. Glutathione S-transferases and their implications in the lung diseases asthma and chronic obstructive pulmonary disease: Early life susceptibility? Redox Biol 2021; 43:101995. [PMID: 33979767 PMCID: PMC8131726 DOI: 10.1016/j.redox.2021.101995] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 01/01/2023] Open
Abstract
Our lungs are exposed daily to airborne pollutants, particulate matter, pathogens as well as lung allergens and irritants. Exposure to these substances can lead to inflammatory responses and may induce endogenous oxidant production, which can cause chronic inflammation, tissue damage and remodeling. Notably, the development of asthma and Chronic Obstructive Pulmonary Disease (COPD) is linked to the aforementioned irritants. Some inhaled foreign chemical compounds are rapidly absorbed and processed by phase I and II enzyme systems critical in the detoxification of xenobiotics including the glutathione-conjugating enzymes Glutathione S-transferases (GSTs). GSTs, and in particular genetic variants of GSTs that alter their activities, have been found to be implicated in the susceptibility to and progression of these lung diseases. Beyond their roles in phase II metabolism, evidence suggests that GSTs are also important mediators of normal lung growth. Therefore, the contribution of GSTs to the development of lung diseases in adults may already start in utero, and continues through infancy, childhood, and adult life. GSTs are also known to scavenge oxidants and affect signaling pathways by protein-protein interaction. Moreover, GSTs regulate reversible oxidative post-translational modifications of proteins, known as protein S-glutathionylation. Therefore, GSTs display an array of functions that impact the pathogenesis of asthma and COPD. In this review we will provide an overview of the specific functions of each class of mammalian cytosolic GSTs. This is followed by a comprehensive analysis of their expression profiles in the lung in healthy subjects, as well as alterations that have been described in (epithelial cells of) asthmatics and COPD patients. Particular emphasis is placed on the emerging evidence of the regulatory properties of GSTs beyond detoxification and their contribution to (un)healthy lungs throughout life. By providing a more thorough understanding, tailored therapeutic strategies can be designed to affect specific functions of particular GSTs.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Marijn Berg
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Caspar H J Schiffers
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Vasili Stylianidis
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Maarten van den Berge
- Pulmonology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Martijn C Nawijn
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
28
|
Zallocco L, Giusti L, Ronci M, Mussini A, Trerotola M, Mazzoni MR, Lucacchini A, Sebastiani L. Salivary Proteome Changes in Response to Acute Psychological Stress Due to an Oral Exam Simulation in University Students: Effect of an Olfactory Stimulus. Int J Mol Sci 2021; 22:4295. [PMID: 33919012 PMCID: PMC8122612 DOI: 10.3390/ijms22094295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 12/11/2022] Open
Abstract
The autonomic nervous system (ANS) plays a crucial role both in acute and chronic psychological stress eliciting changes in many local and systemic physiological and biochemical processes. Salivary secretion is also regulated by ANS. In this study, we explored salivary proteome changes produced in thirty-eight University students by a test stress, which simulated an oral exam. Students underwent a relaxation phase followed by the stress test during which an electrocardiogram was recorded. To evaluate the effect of an olfactory stimulus, half of the students were exposed to a pleasant odor diffused in the room throughout the whole session. Saliva samples were collected after the relaxation phase (T0) and the stress test (T1). State anxiety was also evaluated at T0 and T1. Salivary proteins were separated by two-dimensional electrophoresis, and patterns at different times were compared. Spots differentially expressed were trypsin digested and identified by mass spectrometry. Western blot analysis was used to validate proteomic results. Anxiety scores and heart rate changes indicated that the fake exam induced anxiety. Significant changes of α-amylase, polymeric immunoglobulin receptor (PIGR), and immunoglobulin α chain (IGHA) secretion were observed after the stress test was performed in the two conditions. Moreover, the presence of pleasant odor reduced the acute social stress affecting salivary proteome changes. Therefore, saliva proteomic analysis was a useful approach to evaluate the rapid responses associated to an acute stress test also highlighting known biomarkers.
Collapse
Affiliation(s)
- Lorenzo Zallocco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (L.Z.); (M.R.M.)
| | - Laura Giusti
- School of Pharmacy, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University G. D’Annunzio of Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy;
| | - Andrea Mussini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.S.)
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), University “G. D’Annunzio”, 66100 Chieti, Italy;
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Maria Rosa Mazzoni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (L.Z.); (M.R.M.)
| | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, via Savi, 56126 Pisa, Italy
| | - Laura Sebastiani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.S.)
| |
Collapse
|
29
|
Delmond KA, Delleon H, Goveia RM, Teixeira TM, Abreu DC, Mello-Andrade F, Reis AADS, Silva DDME, Barbosa ADP, Tavares RS, Anunciação CE, Silveira-Lacerda E. Influence of genetic polymorphisms in glutathione-S-transferases gene in response to imatinib among Brazilian patients with chronic myeloid leukemia. Mol Biol Rep 2021; 48:2035-2046. [PMID: 33709282 DOI: 10.1007/s11033-020-06093-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/15/2020] [Indexed: 11/26/2022]
Abstract
Polymorphism in metabolizing enzymes can influence drug response as well as the risk for adverse drug reactions. Nevertheless, there are still few studies analyzing the consequence of polymorphisms for the Glutathione-S-transferases (GST) gene to drug response in chronic myeloid leukemia (CML). This study reports, the influence of GSTP1*B and GSTT1/GSTM1null polymorphisms in response to imatinib in CML patients in a Brazilian population. One hundred thirty-nine CML patients from the Clinical Hospital of Goiânia, Goiás, Brazil, treated with imatinib were enrolled in this study. Genotyping of GSTT1 and GSTM1 genes deletions were performed by qPCR and of GSTP1 gene was performed by RFLP-PCR. The frequency of GSTP1*1B, GSTT1 and GSTM1null polymorphisms were determined for all patients. The influence of each patient's genotypes was analyzed with the patient's response to imatinib treatment. Brazilian CML patients revealed GSTT1 and GSTM1 genes deletions. GSTT1 deletion was found in 19.3% of patients and GSTM1 deletion in 48.7% of patients with CML. GSTT1/GSTM1 deletion was found in 11.7% in Brazilian CML patients. The "G allele" of GSTP1*B, is associated with later cytogenetic response in imatinib therapy. While, the gene presence combined with GG genotype (GSTM1 present/GSTPI-GG) conferred a tend to a later cytogenetic response to patients. GSTP1*B and GSTT1/GSTM1null polymorphisms influence treatment response in CML. Brazilian CML patients presenting GSTP1 AA/AG genotypes alone and in combination with GSTT1 null reach the cytogenetic response faster, while patients presenting GSTP1-GG and GSTMI positive genotypes may take longer to achieve cytogenetic response. As a result, it allows a better prognosis, with the use of an alternative therapy, other than reducing treatment cost.
Collapse
Affiliation(s)
- Kezia Aguiar Delmond
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- College of Goyazes Union, Trindade, Goiás, 75380-000, Brazil
| | - Hugo Delleon
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- Uni-Anhanguera University Center of Goias, Goiânia, Goiás, 74423-115, Brazil
| | - Rebeca Mota Goveia
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Thallita Monteiro Teixeira
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Davi Carvalho Abreu
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Francyelli Mello-Andrade
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás, 74055-110, Brazil
| | - Angela Adamski da Silva Reis
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Daniela de Melo E Silva
- Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | | | | | - Carlos Eduardo Anunciação
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Elisângela Silveira-Lacerda
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil.
| |
Collapse
|
30
|
Jerotic D, Suvakov S, Matic M, Alqudah A, Grieve DJ, Pljesa-Ercegovac M, Savic-Radojevic A, Damjanovic T, Dimkovic N, McClements L, Simic T. GSTM1 Modulates Expression of Endothelial Adhesion Molecules in Uremic Milieu. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6678924. [PMID: 33574979 PMCID: PMC7860968 DOI: 10.1155/2021/6678924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 01/08/2023]
Abstract
Deletion polymorphism of glutathione S-transferase M1 (GSTM1), a phase II detoxification and antioxidant enzyme, increases susceptibility to end-stage renal disease (ESRD) as well as the development of cardiovascular diseases (CVD) among ESRD patients and leads to their shorter cardiovascular survival. The mechanisms by which GSTM1 downregulation contributes to oxidative stress and inflammation in endothelial cells in uremic conditions have not been investigated so far. Therefore, the aim of the present study was to elucidate the effects of GSTM1 knockdown on oxidative stress and expression of a panel of inflammatory markers in human umbilical vein endothelial cells (HUVECs) exposed to uremic serum. Additionally, we aimed to discern whether GSTM1-null genotype is associated with serum levels of adhesion molecules in ESRD patients. HUVECs treated with uremic serum exhibited impaired redox balance characterized by enhanced lipid peroxidation and decreased antioxidant enzyme activities, independently of the GSTM1 knockdown. In response to uremic injury, HUVECs exhibited alteration in the expression of a series of inflammatory cytokines including retinol-binding protein 4 (RBP4), regulated on activation, normal T cell expressed and secreted (RANTES), C-reactive protein (CRP), angiogenin, dickkopf-1 (Dkk-1), and platelet factor 4 (PF4). GSTM1 knockdown in HUVECs showed upregulation of monocyte chemoattractant protein-1 (MCP-1), a cytokine involved in the regulation of monocyte migration and adhesion. These cells also have shown upregulated intracellular and vascular cell adhesion molecules (ICAM-1 and VCAM-1). In accordance with these findings, the levels of serum ICAM-1 and VCAM-1 (sICAM-1 and sVCAM-1) were increased in ESRD patients lacking GSTM1, in comparison with patients with the GSTM1-active genotype. Based on these results, it may be concluded that incubation of endothelial cells in uremic serum induces redox imbalance accompanied with altered expression of a series of cytokines involved in arteriosclerosis and atherosclerosis. The association of GSTM1 downregulation with the altered expression of adhesion molecules might be at least partly responsible for the increased susceptibility of ESRD patients to CVD.
Collapse
Affiliation(s)
- Djurdja Jerotic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Sonja Suvakov
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Marija Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. Box 330127 Zarqa 13133, Jordan
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - David J. Grieve
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Marija Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Tatjana Damjanovic
- Clinical Department for Renal Diseases, Zvezdara University Medical Center, 11000 Belgrade, Serbia
| | - Nada Dimkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Clinical Department for Renal Diseases, Zvezdara University Medical Center, 11000 Belgrade, Serbia
| | - Lana McClements
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 2007, NSW, Australia
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| |
Collapse
|
31
|
Zhang J, Ye ZW, Janssen-Heininger Y, Townsend DM, Tew KD. Development of Telintra as an Inhibitor of Glutathione S-Transferase P. Handb Exp Pharmacol 2021; 264:71-91. [PMID: 32767141 PMCID: PMC8963531 DOI: 10.1007/164_2020_392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Glutathione S-transferase P (GSTP) is a component of a complex series of pathways that provide cellular redox homeostasis. It is an abundant protein in certain tumors and is over-expressed in cancer drug resistance. It has diverse cellular functions that include, thiolase activities with small electrophilic agents or susceptible cysteine residues on the protein to mediate S-glutathionylation, and chaperone binding with select protein kinases. Preclinical and clinical testing of a nanomolar inhibitor of GSTP, TLK199 (Telintra; Ezatiostat) has indicated a role for the enzyme in hematopoiesis and utility for the drug in the treatment of patients with myelodysplastic syndrome.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | | | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
32
|
Chen D, Li J, Jiao F, Wang Q, Li J, Pei Y, Zhao M, Song X, Guo X. ZmACY-1 Antagonistically Regulates Growth and Stress Responses in Nicotiana benthamiana. FRONTIERS IN PLANT SCIENCE 2021; 12:593001. [PMID: 34367193 PMCID: PMC8343404 DOI: 10.3389/fpls.2021.593001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 06/21/2021] [Indexed: 05/17/2023]
Abstract
Aminoacylase-1 is a zinc-binding enzyme that is important in urea cycling, ammonia scavenging, and oxidative stress responses in animals. Aminoacylase-1 (ACY-1) has been reported to play a role in resistance to pathogen infection in the model plant Nicotiana benthamiana. However, little is known about its function in plant growth and abiotic stress responses. In this study, we cloned and analyzed expression patterns of ZmACY-1 in Zea mays under different conditions. We also functionally characterized ZmACY-1 in N. benthamiana. We found that ZmACY-1 is expressed specifically in mature shoots compared with other tissues. ZmACY-1 is repressed by salt, drought, jasmonic acid, and salicylic acid, but is induced by abscisic acid and ethylene, indicating a potential role in stress responses and plant growth. The overexpression of ZmACY-1 in N. benthamiana promoted growth rate by promoting growth-related genes, such as NbEXPA1 and NbEIN2. At the same time, the overexpression of ZmACY-1 in N. benthamiana reduced tolerance to drought and salt stress. With drought and salt stress, the activity of protective enzymes, such as peroxidase (POD), superoxide dismutase (SOD), and catalase (CAT) from micrococcus lysodeikticus was lower; while the content of malondialdehyde (MDA) and relative electrolytic leakage was higher in ZmACY-1 overexpression lines than that in wild-type lines. The results indicate that ZmACY-1 plays an important role in the balance of plant growth and defense and can be used to assist plant breeding under abiotic stress conditions.
Collapse
Affiliation(s)
- Dongbin Chen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
| | - Junhua Li
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
| | - Fuchao Jiao
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
| | - Qianqian Wang
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
| | - Jun Li
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
| | - Yuhe Pei
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
| | - Meiai Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
| | - Xiyun Song
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
- *Correspondence: Xiyun Song,
| | - Xinmei Guo
- Key Laboratory of Qingdao Major Crop Germplasm Resource Innovation and Application, Qingdao, China
- College of Agronomy, Qingdao Agricultural University, Qingdao, China
- Xinmei Guo,
| |
Collapse
|
33
|
Abstract
Drug metabolizing enzymes catalyze the biotransformation of many of drugs and chemicals. The drug metabolizing enzymes are distributed among several evolutionary families and catalyze a range of detoxication reactions, including oxidation/reduction, conjugative, and hydrolytic reactions that serve to detoxify potentially toxic compounds. This detoxication function requires that drug metabolizing enzymes exhibit substrate promiscuity. In addition to their catalytic functions, many drug metabolizing enzymes possess functions unrelated to or in addition to catalysis. Such proteins are termed 'moonlighting proteins' and are defined as proteins with multiple biochemical or biophysical functions that reside in a single protein. This review discusses the diverse moonlighting functions of drug metabolizing enzymes and the roles they play in physiological functions relating to reproduction, vision, cell signaling, cancer, and transport. Further research will likely reveal new examples of moonlighting functions of drug metabolizing enzymes.
Collapse
Affiliation(s)
- Philip G Board
- John Curtin School of Medical Research, ANU College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, New York, NY, USA
| |
Collapse
|
34
|
African Vegetables ( Clerodendrum volibile Leaf and Irvingia gabonensis Seed Extracts) Effectively Mitigate Trastuzumab-Induced Cardiotoxicity in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9535426. [PMID: 33178389 PMCID: PMC7644299 DOI: 10.1155/2020/9535426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Trastuzumab (TZM) is a humanized monoclonal antibody that has been approved for the clinical management of HER2-positive metastatic breast and gastric cancers but its use is limited by its cumulative dose and off-target cardiotoxicity. Unfortunately, till date, there is no approved antidote to this off-target toxicity. Therefore, an acute study was designed at investigating the protective potential and mechanism(s) of CVE and IGE in TZM-induced cardiotoxicity utilizing cardiac enzyme and oxidative stress markers and histopathological endpoints. 400 mg/kg/day CVE and IGE dissolved in 5% DMSO in sterile water were investigated in Wistar rats injected with 2.25 mg/kg/day/i.p. route of TZM for 7 days, using serum cTnI and LDH, complete lipid profile, cardiac tissue oxidative stress markers assays, and histopathological examination of TZM-intoxicated heart tissue. Results showed that 400 mg/kg/day CVE and IGE profoundly attenuated increases in the serum cTnI and LDH levels but caused no significant alterations in the serum lipids and weight gain pattern in the treated rats. CVE and IGE profoundly attenuated alterations in the cardiac tissue oxidative stress markers' activities while improving TZM-associated cardiac histological lesions. These results suggest that CVE and IGE could be mediating its cardioprotection via antioxidant, free radical scavenging, and antithrombotic mechanisms, thus, highlighting the therapeutic potentials of CVE and IGE in the management of TZM-mediated cardiotoxicity.
Collapse
|
35
|
Uppugunduri CRS, Muthukumaran J, Robin S, Santos-Silva T, Ansari M. In silico and in vitro investigations on the protein-protein interactions of glutathione S-transferases with mitogen-activated protein kinase 8 and apoptosis signal-regulating kinase 1. J Biomol Struct Dyn 2020; 40:1430-1440. [PMID: 32996404 DOI: 10.1080/07391102.2020.1827036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Cytosolic glutathione S-transferase (GST) enzymes participate in several cellular processes in addition to facilitating glutathione conjugation reactions that eliminate endogenous and exogenous toxic compounds, especially electrophiles. GSTs are thought to interact with various kinases, resulting in the modulation of apoptotic processes and cellular proliferation. The present research used a combination of in silico and in vitro studies to investigate protein-protein interactions between the seven most abundant cytosolic GSTs-GST alpha-1 (GST-A1), GST alpha-2 (GST-A2), GST mu-1 (GST-M1), GST mu-2 (GST-M2), GST mu-5 (GST-M5), GST theta-1 (GST-T1) and GST pi-1 (GST-P1)-and Mitogen-activated protein kinase 8 (MAPK8) and Apoptosis signal-regulating kinase 1 (ASK1). MAPK8 and ASK1 were chosen as this study's protein interaction partners because of their predominant role in electrophile or cytokine-induced stress-mediated apoptosis, inflammation and fibrosis. The highest degree of sequence homology or sequence similarity was observed in two GST subgroups: the GST-A1, GST-A2 and GST-P1 isoforms constituted subgroup1; the GST-M1, GST-M2 and GST-M5 isoforms constituted subgroup 2. The GST-T1 isoform diverged from these isoforms. In silico investigations revealed that GST-M1 showed a significantly higher binding affinity to MAPK8, and its complex was more structurally stable than the other isoforms, in the order GST-M1 > GST-M5 > GST-P1 > GST-A2 > GST-A1 > GST-M2 > GST-T1. Similarly, GST-A1, GST-P1 and GST-T1 actively interacted with ASK1, and their structural stability was also better, in the order GST-T1 > GST-A1 > GST-P1 > GST-A2 > GST-M5 > GST-M1 > GST-M2. To validate in silico results, we performed in vitro crosslinking and mass spectroscopy experiments. Results indicated that GST-M1 interacted with GST-T1 to form heterodimers and confirmed the predicted interaction between GST-M1 and MAPK8.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chakradhara Rao S Uppugunduri
- Onco-Haematology Unit, Department of Paediatrics, Obstetrics and Gynaecology, Geneva University Hospitals, Geneva, Switzerland.,Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jayaraman Muthukumaran
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal.,Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Shannon Robin
- Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Teresa Santos-Silva
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Marc Ansari
- Onco-Haematology Unit, Department of Paediatrics, Obstetrics and Gynaecology, Geneva University Hospitals, Geneva, Switzerland.,Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Ganesan S, Keating AF. Ovarian mitochondrial and oxidative stress proteins are altered by glyphosate exposure in mice. Toxicol Appl Pharmacol 2020; 402:115116. [PMID: 32634520 PMCID: PMC8500330 DOI: 10.1016/j.taap.2020.115116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022]
Abstract
Glyphosate (GLY) usage for weed control is extensive. To investigate ovarian impacts of chronic GLY exposure, female C57BL6 mice were orally administered saline as vehicle control (CT) or GLY at 0.25 (G0.25), 0.5 (G0.5), 1.0 (G1.0), 1.5 (G1.5), or 2 (G2.0) mg/kg for five days per wk. for 20 wks. Feed intake increased (P < .05) in G1.5 and G2.0 mice and body weight increased (P < .05) in G1.0 mice. There was no impact of GLY on estrous cyclicity, nor did GLY affect circulating levels of 17β-estradiol or progesterone. Exposure to GLY did not impact heart, liver, spleen, kidney or uterus weight. Both ovarian weight and follicle number were increased (P < .05) by G2.0 but not affected at lower GLY concentrations. There were no detectable effects of GLY on ovarian protein abundance of pAKT, AKT, pAKT:AKT, γH2AX, STAR, CYP11A1, HSD3B, CYP19A, ERA or ERB. Increased (P < .05) abundance of ATM protein was observed at G0.25 but not higher GLY doses. A dose-dependent effect (P < .10) of GLY exposure on ovarian protein abundance as quantified by LC-MS/MS was observed (G0.25-4 increased, 19 decreased; G0.5-5 increased, 25 decreased; G1.0-65 increased, 7 decreased; G1.5-145 increased, 2 decreased; G2.0-159 increased, 4 decreased). Pathway analysis was performed using DAVID and identified glutathione metabolism, metabolic and proteasome pathways as GLY exposure targets. These data indicate that chronic low-level exposure to GLY alters the ovarian proteome and may ultimately impact ovarian function.
Collapse
Affiliation(s)
- Shanthi Ganesan
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
37
|
Zhao J, Wang L, Li Y, Zhao W, Kang S. Hypomethylation of the GSTM1 promoter is associated with ovarian endometriosis. Hum Reprod 2020; 34:804-812. [PMID: 30989213 DOI: 10.1093/humrep/dez039] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/16/2019] [Accepted: 02/27/2019] [Indexed: 01/10/2023] Open
Abstract
STUDY QUESTION Is the methylation status of the glutathione S-transferase M1 (GSTM1) promoter region altered in patients with ovarian endometriosis, and does this affect the expression of GSTM1 in their endometrial tissues? SUMMARY ANSWER The promoter region of GSTM1 was significantly hypomethylated in the ectopic and eutopic endometrium of patients with ovarian endometriosis and this was associated with higher expression of GSTM1 mRNA. WHAT IS KNOWN ALREADY GSTM1, a member of the glutathione S-transferase family, is primarily known as a detoxification enzyme, but it has also been shown to negatively regulate apoptosis-related signalling cascades through protein-protein interactions with apoptosis signal-regulating kinase-1. STUDY DESIGN, SIZE, DURATION This is a case-control study between September 2013 and December 2016, involving 65 patients with ovarian endometriosis and 53 women without endometriosis. We analysed the methylation status and expression levels of GSTM1 in the ectopic and eutopic endometrium of patients with ovarian endometriosis and the endometrium of women without endometriosis. In addition, we collected endometrial samples from 12 women without endometriosis for endometrial epithelial cell cultures. PARTICIPANTS/MATERIALS, SETTING, METHODS Methylation levels of the GSTM1 promoter region in the ectopic and eutopic endometrial tissues of patients with ovarian endometriosis and the endometrial tissues of women without endometriosis were analysed by pyrosequencing. The expression of GSTM1 mRNA and protein in endometrial tissues was investigated by RT-qPCR and immunohistochemistry, respectively. Primary cell culture, gene transfection, Cell Counting Kit-8 assay and flow cytometry were used to analyse the effect of GSTM1 on viability and apoptosis in endometrial epithelial cells. MAIN RESULTS AND THE ROLE OF CHANCE Compared with that in the endometrium of women without endometriosis, the GSTM1 promoter region was significantly hypomethylated in the ectopic and eutopic endometrium of patients with ovarian endometriosis. Additionally, GSTM1 mRNA and protein levels were significantly higher in the ectopic and eutopic endometrium than in the control endometrium. Moreover, the methylation levels of the GSTM1 promoter region were significantly negatively correlated with the mRNA expression of GSTM1. Furthermore, in vitro results suggested that the over-expression of GSTM1 could significantly increase viability and inhibit apoptosis in endometrial epithelial cells following hormone treatment and withdrawal. LIMITATIONS, REASONS FOR CAUTION Due to restrictions in the isolation and culture of pure populations of endometrial epithelial cells, as well as limitations in the number of passages possible in primary cells, we could not explore the underlying molecular mechanism by which GSTM1 modulates apoptosis in endometrial cells. WIDER IMPLICATIONS OF THE FINDINGS This study provides new evidence to support the notion that endometriosis may be an epigenetic disease. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from the Natural Science Foundation of Hebei Province (Grant number: H2018206200) and the Department of Education of Hebei Province (Grant number: CXZZBS2017114). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Lixian Wang
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yan Li
- Department of Molecular Biology, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Wei Zhao
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Shan Kang
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, PR China
| |
Collapse
|
38
|
Sathiyanathan P, Samsonraj RM, Tan CLL, Ling L, Lezhava A, Nurcombe V, Stanton LW, Cool SM. A genomic biomarker that identifies human bone marrow-derived mesenchymal stem cells with high scalability. Stem Cells 2020; 38:1124-1136. [PMID: 32510174 DOI: 10.1002/stem.3203] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/29/2020] [Indexed: 12/12/2022]
Abstract
Although the application of human mesenchymal stem cells (hMSCs) to repair damaged or diseased tissues has proven relatively effective, both the donor-to-donor variability in ex vivo expansion rates and the maintenance of stemness remain a bottleneck to widespread translation. Previous work from this laboratory stratified donors into those yielding hMSCs with high- or low-growth capacity; global transcriptomic analysis revealed that high-growth-capacity hMSCs were characterized by a loss of the gene encoding glutathione S-transferase theta 1 (GSTT1). These GSTT1-null hMSCs demonstrated increased proliferative rates, clonogenic potential, and longer telomeres compared with low-growth capacity hMSCs that were GSTT1-positive. Thus, this study identifies GSTT1 as a novel genomic DNA biomarker for hMSC scalability.
Collapse
Affiliation(s)
- Padmapriya Sathiyanathan
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Rebekah M Samsonraj
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Clarissa L L Tan
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Alexander Lezhava
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Lawrence W Stanton
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Simon M Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Orthopaedic Surgery, National University of Singapore, Singapore
| |
Collapse
|
39
|
Llavanera M, Mateo-Otero Y, Bonet S, Barranco I, Fernández-Fuertes B, Yeste M. The triple role of glutathione S-transferases in mammalian male fertility. Cell Mol Life Sci 2020; 77:2331-2342. [PMID: 31807814 PMCID: PMC11105063 DOI: 10.1007/s00018-019-03405-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/11/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Abstract
Male idiopathic infertility accounts for 15-25% of reproductive failure. One of the factors that has been linked to this condition is oxidative stress (OS), defined as the imbalance between antioxidants and reactive oxygen species. Amongst the different factors that protect the cell against OS, the members of the glutathione S-transferase (GST) superfamily play an important role. Interestingly, reduction or lack of some GSTs has been associated to infertility in men. Therefore, and to clarify the relationship between GSTs and male fertility, the aim of this work is to describe the role that GSTs play in the male reproductive tract and in sperm physiology. To that end, the present review provides a novel perspective on the triple role of GSTs (detoxification, regulation of cell signalling and fertilisation), and reports their localisation in sperm, seminal plasma and the male reproductive tract. Furthermore, we also tackle the existing correlation between some GST classes and male fertility. Due to the considerable impact of GSTs in human pathology and their tight relationship with fertility, future research should address the specific role of these proteins in male fertility, which could result in new approaches for the diagnosis and/or treatment of male infertility.
Collapse
Affiliation(s)
- Marc Llavanera
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain
| | - Yentel Mateo-Otero
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain
| | - Sergi Bonet
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain
| | - Isabel Barranco
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain
| | - Beatriz Fernández-Fuertes
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Unit of Cell Biology, Department of Biology, Institute of Food and Agricultural Technology, Faculty of Sciences, University of Girona, C/Maria Aurèlia Campany, 69, Campus Montilivi, 17003, Girona, Spain.
| |
Collapse
|
40
|
Cui Z, Liu Y, Wan W, Xu Y, Hu Y, Ding M, Dou X, Wang R, Li H, Meng Y, Li W, Jiang W, Li Z, Li Y, Tan M, Ma DK, Ding Y, Liu JO, Luo C, Yu B, Tang Q, Dang Y. Ethacrynic acid targets GSTM1 to ameliorate obesity by promoting browning of white adipocytes. Protein Cell 2020; 12:493-501. [PMID: 32399897 PMCID: PMC8160069 DOI: 10.1007/s13238-020-00717-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Zhaomeng Cui
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China.,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200030, China
| | - Wei Wan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yuyan Xu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Yehui Hu
- Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Meng Ding
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China.,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200030, China
| | - Xin Dou
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China.,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200030, China
| | - Ruina Wang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Hailing Li
- Henan Sesame Research Center, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Yongmei Meng
- College of Traditional Mongolian Medicine, Inner Mongolia Medical University, Mongolia, 010110, China
| | - Wei Li
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Wei Jiang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Zengxia Li
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dengke K Ma
- Department of Physiology, Cardiovascular Research Institute, University of California San Francisco, San Francisco, USA
| | - Yu Ding
- Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Qiqun Tang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China. .,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200030, China.
| | - Yongjun Dang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200030, China.
| |
Collapse
|
41
|
Zhang HC, Yang YJ, Ma KX, Shi CY, Chen GW, Liu DZ. A novel sigma class glutathione S-transferase gene in freshwater planarian Dugesia japonica: cloning, characterization and protective effects in herbicide glyphosate stress. ECOTOXICOLOGY (LONDON, ENGLAND) 2020; 29:295-304. [PMID: 32088881 DOI: 10.1007/s10646-020-02173-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/30/2020] [Indexed: 05/27/2023]
Abstract
As the top-selling herbicide in the world, glyphosate distributes widely in natural environment and its influence on the ecological security and human health has attracted more and more concern. Glutathione S-transferases (GSTs) are a well-characterized superfamily of isoenzymes for cellular defense against exogenous toxic substances and therefore protect organisms from injury. In this study, the complete cDNA sequence of GST gene (named as Dja-GST) in freshwater planarian Dugesia japonica was firstly cloned by means of RACE method. The full-length Dja-GST comprises of 706 nucleotides which encodes a polypeptide of 200 amino acids. Dja-GST has two representative GST domains at the N- and C-termini. The conservative GST-N domain includes G-site Y8, F9, R14, W39, K43, P52 and S64, while the variable GST-C domain contains H-site K104, V156, D159 and L161. Sequence analysis, phylogenetic tree reconstruction and multiple alignment collectively indicate that Dja-GST belongs to the Sigma class of GST superfamily. Also, GST gene expression profile, GST enzymatic activity and MDA content in response to glyphosate exposure were systematically investigated and the correlations among them were analyzed. The results suggest that glyphosate exposure modified the mRNA transcription and enzymatic activity of GST, as well as the MDA content in planarians, indicating that Dja-GST might play an important part in organisms defending against oxidative stress induced by glyphosate. This work lays a molecular foundation for further exploring the exact functions of Dja-GST and gives an important implication for evaluating the ecological environment effects of herbicide glyphosate.
Collapse
Affiliation(s)
- He-Cai Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Yu-Juan Yang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Ke-Xue Ma
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Chang-Ying Shi
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Guang-Wen Chen
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China.
| | - De-Zeng Liu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| |
Collapse
|
42
|
Galmés-Pascual BM, Martínez-Cignoni MR, Morán-Costoya A, Bauza-Thorbrügge M, Sbert-Roig M, Valle A, Proenza AM, Lladó I, Gianotti M. 17β-estradiol ameliorates lipotoxicity-induced hepatic mitochondrial oxidative stress and insulin resistance. Free Radic Biol Med 2020; 150:148-160. [PMID: 32105829 DOI: 10.1016/j.freeradbiomed.2020.02.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 02/08/2023]
Abstract
The prevalence and severity of nonalcoholic fatty liver disease (NAFLD) is higher in men and postmenopausal women compared to premenopausal women, suggesting a protective role for ovarian hormones. Diet-induced obesity and fatty acids surplus promote mitochondrial dysfunction in liver, triggering oxidative stress and activation of c-Jun N-terminal kinase (JNK) which has been related to the development of insulin resistance and steatosis, the main hallmarks of NAFLD. Considering that estrogen, in particular 17β-estradiol (E2), have been reported to improve mitochondrial biogenesis and function in liver, our aim was to elucidate the role of E2 in preventing fatty acid-induced insulin resistance in hepatocytes through modulation of mitochondrial function, oxidative stress and JNK activation. An in vivo study was conducted in Wistar rats of both sexes (n = 7) fed control diet and high-fat diet (HFD), and in vitro studies were carried out in HepG2 cells treated with palmitate (PA) and E2 for 24 h. Our HFD-fed male rats showed a prediabetic state characterized by greater systemic and hepatic insulin resistance, as well as higher lipid content in liver, compared to females. JNK activation rose markedly in males in response to HFD feeding, in parallel with mitochondrial dysfunction and oxidative stress. Consistently, in PA-exposed HepG2 cells, E2 treatment prevented JNK activation, insulin resistance and fatty acid accumulation. Altogether, our data highlights the importance of E2 as a mitigating factor of fatty acid-insulin resistance in hepatocytes through downregulation of JNK activation, by means of mitochondrial function improvement.
Collapse
Affiliation(s)
- Bel M Galmés-Pascual
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Melanie Raquel Martínez-Cignoni
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Andrea Morán-Costoya
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain
| | - Marco Bauza-Thorbrügge
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Miquel Sbert-Roig
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Adamo Valle
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, E- 28029, Madrid, Spain.
| | - Ana M Proenza
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, E- 28029, Madrid, Spain
| | - Isabel Lladó
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, E- 28029, Madrid, Spain
| | - Magdalena Gianotti
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa, km 7.5, E-07122, Palma de Mallorca, Illes Balears, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitari Son Espases, E-07120, Palma de Mallorca, Illes Balears, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, E- 28029, Madrid, Spain
| |
Collapse
|
43
|
Drug-metabolizing enzymes: role in drug resistance in cancer. Clin Transl Oncol 2020; 22:1667-1680. [PMID: 32170639 DOI: 10.1007/s12094-020-02325-7] [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: 11/01/2019] [Accepted: 02/18/2020] [Indexed: 12/22/2022]
Abstract
Although continuous researches are going on for the discovery of new chemotherapeutic agents, resistance to these anticancer agents has made it really difficult to reach the fruitful results. There are many causes for this resistance that are being studied by the researchers across the world, but still, success is far because there are several factors that are going along unattended or have been studied less. Drug-metabolizing enzymes (DMEs) are one of these factors, on which less study has been conducted. DMEs include Phase I and Phase II enzymes. Cytochrome P450s (CYPs) are major Phase I enzymes while glutathione-S-transferases (GSTs), UDP-glucuronosyltransferases (UGTs), dihydropyrimidine dehydrogenases are the major enzymes belonging to the Phase II enzymes. These enzymes play an important role in detoxification of the xenobiotics as well as the metabolism of drugs, depending upon the tissue in which they are expressed. When present in tumorous tissues, they cause resistance by metabolizing the drugs and rendering them inactive. In this review, the role of these various enzymes in anticancer drug metabolism and the possibilities for overcoming the resistance have been discussed.
Collapse
|
44
|
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
|
45
|
Gigliotti JC, Tin A, Pourafshar S, Cechova S, Wang YT, Sung SSJ, Bodonyi-Kovacs G, Cross JV, Yang G, Nguyen N, Chan F, Rebholz C, Yu B, Grove ML, Grams ME, Köttgen A, Scharpf R, Ruiz P, Boerwinkle E, Coresh J, Le TH. GSTM1 Deletion Exaggerates Kidney Injury in Experimental Mouse Models and Confers the Protective Effect of Cruciferous Vegetables in Mice and Humans. J Am Soc Nephrol 2020; 31:102-116. [PMID: 31727850 PMCID: PMC6935006 DOI: 10.1681/asn.2019050449] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/07/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND GSTM1 encodes glutathione S-transferase μ-1 (GSTM1), which belongs to a superfamily of phase 2 antioxidant enzymes. The highly prevalent GSTM1 deletion variant is associated with kidney disease progression in human cohorts: the African American Study of Kidney Disease and Hypertension and the Atherosclerosis Risk in Communities (ARIC) Study. METHODS We generated a Gstm1 knockout mouse line to study its role in a CKD model (involving subtotal nephrectomy) and a hypertension model (induced by angiotensin II). We examined the effect of intake of cruciferous vegetables and GSTM1 genotypes on kidney disease in mice as well as in human ARIC study participants. We also examined the importance of superoxide in the mediating pathways and of hematopoietic GSTM1 on renal inflammation. RESULTS Gstm1 knockout mice displayed increased oxidative stress, kidney injury, and inflammation in both models. The central mechanism for kidney injury is likely mediated by oxidative stress, because treatment with Tempol, an superoxide dismutase mimetic, rescued kidney injury in knockout mice without lowering BP. Bone marrow crosstransplantation revealed that Gstm1 deletion in the parenchyma, and not in bone marrow-derived cells, drives renal inflammation. Furthermore, supplementation with cruciferous broccoli powder rich in the precursor to antioxidant-activating sulforaphane significantly ameliorated kidney injury in Gstm1 knockout, but not wild-type mice. Similarly, among humans (ARIC study participants), high consumption of cruciferous vegetables was associated with fewer kidney failure events compared with low consumption, but this association was observed primarily in participants homozygous for the GSTM1 deletion variant. CONCLUSIONS Our data support a role for the GSTM1 enzyme in the modulation of oxidative stress, inflammation, and protective metabolites in CKD.
Collapse
Affiliation(s)
| | - Adrienne Tin
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | | | | | - Yves T Wang
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York
| | | | | | - Janet V Cross
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Guang Yang
- Division of Nephrology, Heinrich-Heine University of Dusseldorf, Dusseldorf, Germany
| | - Nhu Nguyen
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan
| | - Fang Chan
- Division of Nephrology, Department of Medicine and
| | - Casey Rebholz
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health and
| | - Megan L Grove
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Morgan E Grams
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
- Department of Medicine and
| | - Anna Köttgen
- Department of Epidemiology and
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; and
| | - Robert Scharpf
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland
- Division of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Phillip Ruiz
- Department of Pathology, University of Miami, Miami, Florida
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Josef Coresh
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | - Thu H Le
- Division of Nephrology, Department of Medicine and
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland
| |
Collapse
|
46
|
Cloning, characterization and expression analysis of glutathione S-transferase from the Antarctic yeast Rhodotorula mucilaginosa AN5. Protein Expr Purif 2019; 167:105518. [PMID: 31669543 DOI: 10.1016/j.pep.2019.105518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
The gene for glutathione S-transferase (GST) in Antarctic sea-ice yeast Rhodotorula mucilaginosa AN5 was cloned and expressed in Escherichia coli and named RmGST. Sequence analysis showed that the RmGST gene contained a 843 bp open reading frame, which encoded 280 amino acid residues with a calculated molecular mass of 30.4 kDa and isoelectric point of 5.40. RmGST has the typical C- and N-terminal double domains of glutathione S-transferase. Recombinant RmGST (rRmGST) was expressed in E. coli to produce heterologous protein that had a high specific activity of 60.2 U/mg after purification. The apparent Km values of rRmGST for glutathione and 1-chloro-2,4-dinitrobenzene were 0.35 mM and 0.40 mM, respectively. Optimum enzyme activity was measured at 35 °C and at pH 7.0 and complete inactivation was observed after incubation at 55 °C for 60 min rRmGST tolerated high salt concentrations (1.0 M NaCl) and was stable at pH 3.0. Additionally, the recombinant protein nearly kept whole activity in Hg2+ and Mn2+, and could tolerate Ca2+, Cu2+, Mg2+, Cd2+, EDTA, thiourea, urea, Tween-80, H2O2 and Triton X-100. Real-time quantitative PCR showed that relative expression of the GST gene was significantly increased under Cu2+ and low temperature stress. These results indicate that rRmGST is a typical low thermostable enzyme, while its other characteristics, heavy metal and low temperature tolerance, might be related to its Antarctic home environment.
Collapse
|
47
|
Liu K, Singer E, Cohn W, Micewicz ED, McBride WH, Whitelegge JP, Loo JA. Time-Dependent Measurement of Nrf2-Regulated Antioxidant Response to Ionizing Radiation Toward Identifying Potential Protein Biomarkers for Acute Radiation Injury. Proteomics Clin Appl 2019; 13:e1900035. [PMID: 31419066 PMCID: PMC7213060 DOI: 10.1002/prca.201900035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/16/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE Potential acute exposure to ionizing radiation in nuclear or radiological accidents presents complex mass casualty scenarios that demand prompt triage and treatment decisions. Due to delayed symptoms and varied response of radiation victims, there is an urgent need to develop robust biomarkers to assess the extent of injuries in individuals. EXPERIMENTAL DESIGN The transcription factor Nrf2 is the master of redox homeostasis and there is transcriptional evidence of Nrf2-dependent antioxidant response activation upon radiation. The biomarker potential of Nrf2-dependent downstream target enzymes is investigated by measuring their response in bone marrow extracted from C57Bl/6 and C3H mice of both genders for up to 4 days following 6 Gy total body irradiation using targeted MS. RESULTS Overall, C57Bl/6 mice have a stronger proteomic response than C3H mice. In both strains, male mice have more occurrences of upregulation in antioxidant enzymes than female mice. For C57Bl/6 male mice, three proteins show elevated abundances after radiation exposure: catalase, superoxide dismutase 1, and heme oxygenase 1. Across both strains and genders, glutathione S-transferase Mu 1 is consistently decreased. CONCLUSIONS AND CLINICAL RELEVANCE This study provides the basis for future development of organ-specific protein biomarkers used in diagnostic blood test for radiation injury.
Collapse
Affiliation(s)
- Kate Liu
- Department of Chemistry and Biochemistry, UCLA
| | - Elizabeth Singer
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA
| | - Whitaker Cohn
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA
| | - Ewa D. Micewicz
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA
| | | | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, UCLA
- Department of Biological Chemistry, David Geffen School of Medicine, Molecular Biology Institute, and UCLA/DOE Institute for Genomics and Proteomics, UCLA
| |
Collapse
|
48
|
Taeb M, Jafarzadeh A, Mortazavi-Jahromi SS, Zainodini N, Mirzaei MR, Jafarnezhad-Ansariha F, Aghazadeh Z, Mirshafiey A. Effect of β-D-Mannuronic Acid (M2000) on Oxidative Stress Enzymes' Gene Using Healthy Donor Peripheral Blood Mononuclear Cells for Evaluating the Anti-Aging Property. Curr Drug Discov Technol 2019; 16:265-271. [PMID: 29766814 DOI: 10.2174/1570163815666180515122834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/07/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This research aimed to study the anti-aging and anti-inflammatory effects of low and high doses of the β-D-mannuronic (M2000) on gene expression of enzymes involved in oxidative stress (including SOD2, GST, GPX1, CAT, iNOS, and MPO) in peripheral blood mononuclear cells (PBMCs) of healthy donors under in vitro conditions. METHODS The PBMCs were separated and the RNAs were then extracted and the cDNAs synthesized, and expression levels of the mentioned genes were detected by qRT-PCR. RESULTS Our results indicated that the high dose of this drug could significantly reduce the expression level of the SOD2 gene compared to the lipopolysaccharide (LPS) group (p < 0.0001). Moreover, it was found that the high dose of this drug could significantly decrease the expression level of the GST gene compared to the LPS group (p < 0.0001). However, no significant reductions were observed in expression levels of the CAT and GPX1 genes compared to the LPS group. Furthermore, our data revealed that the level of iNOS and MPO gene expression was significantly reduced, in both doses of M2000, respectively, compared to the LPS group (p < 0.0001). CONCLUSION This research showed that M2000 as a novel NSAID with immunosuppressive properties could modify oxidative stress through lowering expression levels of the SOD2, GST, iNOS, and MPO genes compared to the healthy expression levels, with a probable reduction of the risk of developing inflammatory diseases related to age and aging.
Collapse
Affiliation(s)
- Mahsa Taeb
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Seyed Shahabeddin Mortazavi-Jahromi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Department of Cellular and Molecular Biology, Kish International Campus, University of Tehran, Kish, Iran
| | - Nahid Zainodini
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Mirzaei
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Zahra Aghazadeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Samaraweera AV, Sandamalika WMG, Liyanage DS, Lee S, Priyathilaka TT, Lee J. Molecular characterization and functional analysis of glutathione S-transferase kappa 1 (GSTκ1) from the big belly seahorse (Hippocampus abdominalis): Elucidation of its involvement in innate immune responses. FISH & SHELLFISH IMMUNOLOGY 2019; 92:356-366. [PMID: 31200074 DOI: 10.1016/j.fsi.2019.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/03/2019] [Accepted: 06/09/2019] [Indexed: 06/09/2023]
Abstract
Glutathione S-transferases (GSTs) are essential enzymes for the bioactivation of xenobiotics through the conjugation of the thiol group of glutathione (GSH). In this study, a kappa class of GST was identified from the big belly seahorse (Hippocampus abdominalis) (HaGSTκ1) and its biochemical and functional properties were analyzed. HaGSTκ1 has 231 amino acids encoded by a 696 bp open reading frame (ORF). The protein has a predicted molecular mass of 26.04 kDa and theoretical isoelectric point (pI) of 8.28. It comprised a thioredoxin domain, disulfide bond formation protein A (DsbA) general fold, and Ser15 catalytic site as well as GSH-binding and polypeptide-binding sites. Phylogenetic analysis revealed that HaGSTκ1 is closely clustered with the kappa class of GSTs from teleost fishes. The recombinant (rHaGSTκ1) protein exhibited activity toward 1-chloro-2,4-dinitrobenzene (CDNB), 4-nitrobenzyl (4-NBC), and 4-nitrophenethyl bromide (4-NPB) but not 1,2-dichloro-4-nitrobenzene (DCNB). The optimum pH and temperature were 8 and 30 °C, respectively, for the catalysis of CDNB and the universal substrate of GSTs. The rHaGSTκ1 activity was efficiently inhibited in the presence of Cibacron blue (CB) as compared with hematin. Most prominent expression of HaGSTκ1 was observed in the liver and kidney among the fourteen different tissues of normal seahorse. After challenge with lipopolysaccharide (LPS), polyinosinic-polycytidylic (poly I:C), gram-negative Edwardsiella tarda, and gram-positive Streptococcus iniae, HaGSTκ1 expression was significantly modulated in the liver and blood tissues. Altogether, our study proposes the plausible important role of HaGSTκ1 in innate immunity and detoxification of harmful xenobiotics.
Collapse
Affiliation(s)
- Anushka Vidurangi Samaraweera
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - W M Gayashani Sandamalika
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Sukkyoung Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Thanthrige Thiunuwan Priyathilaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
50
|
Ji Y, Dai F, Yan S, Shi JY, Zhou B. Identification of Catechol-Type Diphenylbutadiene as a Tyrosinase-Activated Pro-oxidative Chemosensitizer against Melanoma A375 Cells via Glutathione S-Transferase Inhibition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9060-9069. [PMID: 31339696 DOI: 10.1021/acs.jafc.9b02875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Glutathione S-transferases (GSTs) play an active role in the development of drug resistance by numerous cancer cells, including melanoma cells, which is a major cause of chemotherapy failure. As part of our continuous effort to explore why dietary polyphenols bearing the catechol moiety (dietary catechols) show usually anticancer activity, catechol-type diphenylbutadiene (3,4-DHB) was selected as a model of dietary catechols to probe whether they work as pro-oxidative chemosensitizers via GST inhibition in melanoma cells. It was found that, in human melanoma A375 cells, 3,4-DHB is easily converted to its ortho-quinone via copper-containing tyrosinase-mediated two-electron oxidation along with generation of reactive oxygen species (ROS) derived from the oxidation; the resulting ortho-quinone and ROS are responsible for its ability to sensitize the cisplatin-resistant cells by inhibiting GST, followed by induction of apoptosis in an ASK1-JNK/p38 signaling cascade and mitochondria-dependent pathway. This work provides further evidence to support that dietary catechols exhibit antimelanoma activity by virtue of their tyrosinase-dependent pro-oxidative role and gives useful information for designing polyphenol-inspired GST inhibitors and sensitizers in chemotherapy against melanoma.
Collapse
Affiliation(s)
- Yuan Ji
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , People's Republic of China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , People's Republic of China
| | - Shuai Yan
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , People's Republic of China
| | - Jing-Yang Shi
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , People's Republic of China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , People's Republic of China
| |
Collapse
|