51
|
Wang WW, Wang J, Zhang HJ, Wu SG, Qi GH. Transcriptome analysis reveals mechanism underlying the differential intestinal functionality of laying hens in the late phase and peak phase of production. BMC Genomics 2019; 20:970. [PMID: 31830910 PMCID: PMC6907226 DOI: 10.1186/s12864-019-6320-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Background The compromised performance of laying hens in the late phase of production relative to the peak production was thought to be associated with the impairment of intestinal functionality, which plays essential roles in contributing to their overall health and production performance. In the present study, RNA sequencing was used to investigate differences in the expression profile of intestinal functionality-related genes and associated pathways between laying hens in the late phase and peak phase of production. Results A total of 104 upregulated genes with 190 downregulated genes were identified in the ileum (the distal small intestine) of laying hens in the late phase of production compared to those at peak production. These upregulated genes were found to be enriched in little KEGG pathway, however, the downregulated genes were enriched in the pathways of PPAR signaling pathway, oxidative phosphorylation and glutathione metabolism. Besides, these downregulated genes were mapped to several GO clusters in relation to lipid metabolism, electron transport of respiratory chain, and oxidation resistance. Similarly, there were lower activities of total superoxide dismutase, glutathione S-transferase and Na+/K+-ATPase, and reductions of total antioxidant capacity and ATP level, along with an elevation in malondialdehyde content in the ileum of laying hens in the late phase of production as compared with those at peak production. Conclusions The intestine of laying hens in the late phase of production were predominantly characterized by a disorder of lipid metabolism, concurrent with impairments of energy production and antioxidant property. This study uncovers the mechanism underlying differences between the intestinal functionality of laying hens in the late phase and peak phase of production, thereby providing potential targets for the genetic control or dietary modulation of intestinal hypofunction of laying hens in the late phase of production.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture & Rural Affairs, National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Jing Wang
- Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture & Rural Affairs, National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Hai-Jun Zhang
- Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture & Rural Affairs, National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Shu-Geng Wu
- Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture & Rural Affairs, National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Guang-Hai Qi
- Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture & Rural Affairs, National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
| |
Collapse
|
52
|
Glutathione Transferase Omega-1 Regulates NLRP3 Inflammasome Activation through NEK7 Deglutathionylation. Cell Rep 2019; 29:151-161.e5. [DOI: 10.1016/j.celrep.2019.08.072] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 07/31/2019] [Accepted: 08/23/2019] [Indexed: 01/20/2023] Open
|
53
|
Ahuié Kouakou G, Gagnon H, Lacasse V, Wagner JR, Naylor S, Klarskov K. Dehydroascorbic acid S-Thiolation of peptides and proteins: Role of homocysteine and glutathione. Free Radic Biol Med 2019; 141:233-243. [PMID: 31228548 DOI: 10.1016/j.freeradbiomed.2019.06.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 01/05/2023]
Abstract
Ascorbic acid (vitamin C) plays a significant role in the prevention of oxidative stress. In this process, ascorbate is oxidized to dehydroascorbate (DHA). We have investigated the impact of DHA on peptide/protein intramolecular disulfide formation as well as S-glutathionylation and S-homocysteinylation. S-glutathionylation of peptides/proteins is a reversible, potential regulatory mechanism in oxidative stress. Although the exact role of protein S-homocysteinylation is unknown, it has been proposed to be of importance in pathobiological processes such as onset of cardiovascular disease. Using an in vitro model system, we demonstrate that DHA causes disulfide bond formation within the active site of recombinant human glutaredoxin (Grx-1). DHA also facilities the formation of S-glutathionylation and S-homocysteinylation of a model peptide (AcFHACAAK) as well as Grx-1. We discuss the possible mechanisms of peptide/protein S-thiolation, which can occur either via thiol exchange or a thiohemiketal intermediate. A thiohemiketal DHA-peptide adduct was detected by mass spectrometry and its location on the peptide/protein cysteinyl thiol group was unambiguously confirmed by tandem mass spectrometry. This demonstrates that peptide/protein S-thiolation mediated by DHA is not limited to thiol exchange reactions but also takes place directly via the formation of a thiohemiketal peptide intermediate. Finally, we investigated a potential reducing role of glutathione (GSH) in the presence of S-homocysteinylated peptide/protein adducts. S-homocysteinylated AcFHACAAK, human hemoglobin α-chain and Grx-1 were incubated with GSH. Both peptide and proteins were reduced, and homocysteine replaced with GS-adducts by thiol exchange, as a function of time.
Collapse
Affiliation(s)
- Grace Ahuié Kouakou
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Canada
| | - Hugo Gagnon
- PhenoSwitch Bioscience, 975 Rue Léon-Trépanier, Sherbrooke, QC J1G 5J6, Canada
| | - Vincent Lacasse
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Canada
| | - J Richard Wagner
- Département de Médecine Nucléaire et radiobiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Canada
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Drive, Elm Grove, WI, 53122, USA
| | - Klaus Klarskov
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Canada.
| |
Collapse
|
54
|
Fernando N, Wooff Y, Aggio-Bruce R, Chu-Tan JA, Jiao H, Dietrich C, Rutar M, Rooke M, Menon D, Eells JT, Valter K, Board PG, Provis J, Natoli R. Photoreceptor Survival Is Regulated by GSTO1-1 in the Degenerating Retina. Invest Ophthalmol Vis Sci 2019; 59:4362-4374. [PMID: 30193308 DOI: 10.1167/iovs.18-24627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Glutathione-S-transferase omega 1-1 (GSTO1-1) is a cytosolic glutathione transferase enzyme, involved in glutathionylation, toll-like receptor signaling, and calcium channel regulation. GSTO1-1 dysregulation has been implicated in oxidative stress and inflammation, and contributes to the pathogenesis of several diseases and neurological disorders; however, its role in retinal degenerations is unknown. The aim of this study was to investigate the role of GSTO1-1 in modulating oxidative stress and consequent inflammation in the normal and degenerating retina. Methods The role of GSTO1-1 in retinal degenerations was explored by using Gsto1-/- mice in a model of retinal degeneration. The expression and localization of GSTO1-1 were investigated with immunohistochemistry and Western blot. Changes in the expression of inflammatory (Ccl2, Il-1β, and C3) and oxidative stress (Nox1, Sod2, Gpx3, Hmox1, Nrf2, and Nqo1) genes were investigated via quantitative real-time polymerase chain reaction. Retinal function in Gsto1-/- mice was investigated by using electroretinography. Results GSTO1-1 was localized to the inner segment of cone photoreceptors in the retina. Gsto1-/- photo-oxidative damage (PD) mice had decreased photoreceptor cell death as well as decreased expression of inflammatory (Ccl2, Il-1β, and C3) markers and oxidative stress marker Nqo1. Further, retinal function in the Gsto1-/- PD mice was increased as compared to wild-type PD mice. Conclusions These results indicate that GSTO1-1 is required for inflammatory-mediated photoreceptor death in retinal degenerations. Targeting GSTO1-1 may be a useful strategy to reduce oxidative stress and inflammation and ameliorate photoreceptor loss, slowing the progression of retinal degenerations.
Collapse
Affiliation(s)
- Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.,The ANU Medical School, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Joshua A Chu-Tan
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Haihan Jiao
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Catherine Dietrich
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Matt Rutar
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Melissa Rooke
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Deepthi Menon
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Janis T Eells
- Department of Biomedical Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States
| | - Krisztina Valter
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.,The ANU Medical School, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Philip G Board
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jan Provis
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.,The ANU Medical School, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.,The ANU Medical School, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
55
|
Manupati K, Debnath S, Goswami K, Bhoj PS, Chandak HS, Bahekar SP, Das A. Glutathione S-transferase omega 1 inhibition activates JNK-mediated apoptotic response in breast cancer stem cells. FEBS J 2019; 286:2167-2192. [PMID: 30873742 DOI: 10.1111/febs.14813] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 11/13/2018] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
Glutathione S-transferase omega 1 (GSTO1) contributes to the inactivation of a wide range of drug compounds via conjugation to glutathione during phase reactions. Chemotherapy-induced GSTO1 expression in breast cancer cells leads to chemoresistance and promotes metastasis. In search of novel GSTO1 inhibitors, we identified S2E, a thia-Michael adduct of sulfonamide chalcone with low LC50 (3.75 ± 0.73 μm) that binds to the active site of GSTO1, as revealed by molecular docking (glide score: -8.1), cellular thermal shift assay and fluorescence quenching assay (Kb ≈ 10 × 105 mol·L-1 ). Docking studies confirmed molecular interactions between GSTO1 and S2E, and identified the hydrogen bond donor Val-72 (2.14 Å) and hydrogen bond acceptor Ser-86 (2.77 Å). Best pharmacophore hypotheses could effectively map S2E and identified the 4-methyl group of the benzene sulfonamide ring as crucial to its anti-cancer activity. Lack of a thiophenyl group in another analog, 2e, reduced its efficacy as observed by cytotoxicity and pharmacophore matching. Furthermore, GSTO1 inhibition by S2E, along with tamoxifen, led to a significant increase in apoptosis and decreased migration of aggressive MDA-MB-231 cells, as well as significantly decreased migration, invasion and mammosphere formation in sorted breast cancer stem cells (CSCs, CD24- /CD44+ ). GSTO1 silencing in breast CSCs also significantly increased apoptosis and decreased migration. Mechanistically, GSTO1 inhibition activated the c-Jun N-terminal kinase stress kinase, inducing a mitochondrial apoptosis signaling pathway in breast CSCs via the pro-apoptotic proteins BAX, cytochrome c and cleaved caspase 3. Our study elucidated the role of the GSTO1 inhibitor S2E as a potential therapeutic strategy for preventing chemotherapy-induced breast CSC-mediated cancer metastasis and recurrence.
Collapse
Affiliation(s)
- Kanakaraju Manupati
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Science and Innovative Research, New Delhi, India
| | - Sudhan Debnath
- Department of Chemistry, Maharaja Bir Bikram College, Agartala, India
| | - Kalyan Goswami
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Priyanka S Bhoj
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Hemant S Chandak
- Department of Chemistry, G. S. Science, Arts & Commerce College, Khamgaon, India
| | - Sandeep P Bahekar
- Department of Chemistry, G. S. Science, Arts & Commerce College, Khamgaon, India
| | - Amitava Das
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Science and Innovative Research, New Delhi, India
| |
Collapse
|
56
|
Saisawang C, Wongsantichon J, Robinson RC, Ketterman AJ. Glutathione transferase Omega 1‐1 (GSTO1‐1) modulates Akt and MEK1/2 signaling in human neuroblastoma cell SH‐SY5Y. Proteins 2019; 87:588-595. [DOI: 10.1002/prot.25683] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/21/2019] [Accepted: 03/13/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Chonticha Saisawang
- Institute of Molecular BiosciencesMahidol University Salaya Nakhon Pathom Thailand
| | - Jantana Wongsantichon
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR) Singapore Singapore
- Mahidol‐Oxford Tropical Medicine Research Unit (MORU) Bangkok Thailand
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR) Singapore Singapore
- Research Institute for Interdisciplinary ScienceOkayama University Okayama Japan
| | - Albert J. Ketterman
- Institute of Molecular BiosciencesMahidol University Salaya Nakhon Pathom Thailand
| |
Collapse
|
57
|
Bartolini D, Torquato P, Piroddi M, Galli F. Targeting glutathione S-transferase P and its interactome with selenium compounds in cancer therapy. Biochim Biophys Acta Gen Subj 2019; 1863:130-143. [DOI: 10.1016/j.bbagen.2018.09.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022]
|
58
|
Sylvestre-Gonon E, Law SR, Schwartz M, Robe K, Keech O, Didierjean C, Dubos C, Rouhier N, Hecker A. Functional, Structural and Biochemical Features of Plant Serinyl-Glutathione Transferases. FRONTIERS IN PLANT SCIENCE 2019; 10:608. [PMID: 31191562 PMCID: PMC6540824 DOI: 10.3389/fpls.2019.00608] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/25/2019] [Indexed: 05/04/2023]
Abstract
Glutathione transferases (GSTs) belong to a ubiquitous multigenic family of enzymes involved in diverse biological processes including xenobiotic detoxification and secondary metabolism. A canonical GST is formed by two domains, the N-terminal one adopting a thioredoxin (TRX) fold and the C-terminal one an all-helical structure. The most recent genomic and phylogenetic analysis based on this domain organization allowed the classification of the GST family into 14 classes in terrestrial plants. These GSTs are further distinguished based on the presence of the ancestral cysteine (Cys-GSTs) present in TRX family proteins or on its substitution by a serine (Ser-GSTs). Cys-GSTs catalyze the reduction of dehydroascorbate and deglutathionylation reactions whereas Ser-GSTs catalyze glutathione conjugation reactions and eventually have peroxidase activity, both activities being important for stress tolerance or herbicide detoxification. Through non-catalytic, so-called ligandin properties, numerous plant GSTs also participate in the binding and transport of small heterocyclic ligands such as flavonoids including anthocyanins, and polyphenols. So far, this function has likely been underestimated compared to the other documented roles of GSTs. In this review, we compiled data concerning the known enzymatic and structural properties as well as the biochemical and physiological functions associated to plant GSTs having a conserved serine in their active site.
Collapse
Affiliation(s)
- Elodie Sylvestre-Gonon
- Interactions Arbres-Microorganismes, Institut National de la Recherche Agronomique, Université de Lorraine, Nancy, France
| | - Simon R. Law
- Department of Plant Physiology, Umeå Plant Science Centre, Umeå University, Umeå, Sweden
| | - Mathieu Schwartz
- Centre National de la Recherche Scientifique, Cristallographie, Résonance Magnétique et Modélisations, Université de Lorraine, Nancy, France
| | - Kevin Robe
- Biochimie et Physiologie Moléculaire des Plantes (BPMP), INRA, CNRS, SupAgro-M, Université de Montpellier, Montpellier, France
| | - Olivier Keech
- Department of Plant Physiology, Umeå Plant Science Centre, Umeå University, Umeå, Sweden
| | - Claude Didierjean
- Centre National de la Recherche Scientifique, Cristallographie, Résonance Magnétique et Modélisations, Université de Lorraine, Nancy, France
| | - Christian Dubos
- Biochimie et Physiologie Moléculaire des Plantes (BPMP), INRA, CNRS, SupAgro-M, Université de Montpellier, Montpellier, France
| | - Nicolas Rouhier
- Interactions Arbres-Microorganismes, Institut National de la Recherche Agronomique, Université de Lorraine, Nancy, France
- *Correspondence: Nicolas Rouhier, Arnaud Hecker,
| | - Arnaud Hecker
- Interactions Arbres-Microorganismes, Institut National de la Recherche Agronomique, Université de Lorraine, Nancy, France
- *Correspondence: Nicolas Rouhier, Arnaud Hecker,
| |
Collapse
|
59
|
Radic TM, Coric VM, Pljesa-Ercegovac MS, Basta-Jovanovic GM, Radojevic-Skodric SM, Dragicevic DP, Matic MG, Bogdanovic LM, Dzamic ZM, Simic TP, Savic-Radojevic AR. Concomitance of Polymorphisms in Glutathione Transferase Omega Genes Is Associated with Risk of Clear Cell Renal Cell Carcinoma. TOHOKU J EXP MED 2018; 246:35-44. [PMID: 30224590 DOI: 10.1620/tjem.246.35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glutathione S-transferases (GSTs), a superfamily of multifunctional enzymes, play an important role in the onset and progression of renal cell carcinoma (RCC). However, novel GST omega class (GSTO), consisting of GSTO1-1 and GSTO2-2 isoenzymes, has not been studied in RCC yet. Two coding single nucleotide polymorphisms (SNPs) supposedly affect their functions: GSTO1*C419A (rs4925) causing alanine to aspartate substitution (*A140D) and GSTO2*A424G (rs156697) causing asparagine to aspartate substitution (*N142D), and have been associated with several neurodegenerative diseases and cancers. Functional relevance of yet another GSTO2 polymorphism, identified at the 5' untranslated (5'UTR) gene region (GSTO2*A183G, rs2297235), has not been clearly discerned so far. Therefore, we aimed to assess the effect of specific GSTO1 and GSTO2 gene variants, independently and in interaction with established risk factors (smoking, obesity and hypertension) on the risk for the most aggressive RCC subtype, the clear cell RCC (ccRCC). Genotyping was performed in 239 ccRCC patients and 350 matched controls, while plasma levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a biomarker of oxidative DNA damage, were determined by ELISA. As a result, combined effect of all three variant genotypes exhibited almost 3-fold risk of RCC development. Additionally, this association was confirmed at the haplotype level [variant GSTO1*A/GSTO2*G (rs156697)/GSTO2*G (rs2297235) haplotype], suggesting a potential role of those variants in propensity to RCC. Regarding the gene-environment interactions, variant GSTO2*G (rs156697) homozygous smokers are at higher ccRCC risk. Association in terms of oxidative DNA damage was found for GSTO2 polymorphism in 5'UTR and 8-OHdG. In conclusion, the concomitance of GSTO polymorphisms may influence ccRCC risk.
Collapse
Affiliation(s)
- Tanja M Radic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Vesna M Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Marija S Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Gordana M Basta-Jovanovic
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Sanja M Radojevic-Skodric
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Dejan P Dragicevic
- Faculty of Medicine, University of Belgrade.,Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade
| | - Marija G Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Ljiljana M Bogdanovic
- Faculty of Medicine, University of Belgrade.,Institute of Pathology, Faculty of Medicine, University of Belgrade
| | - Zoran M Dzamic
- Faculty of Medicine, University of Belgrade.,Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade
| | - Tatjana P Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| | - Ana R Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade.,Faculty of Medicine, University of Belgrade
| |
Collapse
|
60
|
Glutathione Transferases: Potential Targets to Overcome Chemoresistance in Solid Tumors. Int J Mol Sci 2018; 19:ijms19123785. [PMID: 30487385 PMCID: PMC6321424 DOI: 10.3390/ijms19123785] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 12/14/2022] Open
Abstract
Multifunctional enzymes glutathione transferases (GSTs) are involved in the development of chemoresistance, thus representing a promising target for a novel approach in cancer treatment. This superfamily of polymorphic enzymes exhibits extraordinary substrate promiscuity responsible for detoxification of numerous conventional chemotherapeutics, at the same time regulating signaling pathways involved in cell proliferation and apoptosis. In addition to upregulated GST expression, different cancer cell types have a unique GST signature, enabling targeted selectivity for isoenzyme specific inhibitors and pro-drugs. As a result of extensive research, certain GST inhibitors are already tested in clinical trials. Catalytic properties of GST isoenzymes are also exploited in bio-activation of specific pro-drugs, enabling their targeted accumulation in cancer cells with upregulated expression of the appropriate GST isoenzyme. Moreover, the latest approach to increase specificity in treatment of solid tumors is development of GST pro-drugs that are derivatives of conventional anti-cancer drugs. A future perspective is based on the design of new drugs, which would selectively target GST overexpressing cancers more prone to developing chemoresistance, while decreasing side effects in off-target cells.
Collapse
|
61
|
Sadeghi H, Golalipour M, Yamchi A, Farazmandfar T, Shahbazi M. CDC25A pathway toward tumorigenesis: Molecular targets of CDC25A in cell-cycle regulation. J Cell Biochem 2018; 120:2919-2928. [PMID: 30443958 DOI: 10.1002/jcb.26838] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/22/2018] [Indexed: 11/10/2022]
Abstract
The cell division cycle 25 (CDC25) phosphatases regulate key transitions between cell-cycle phases during normal cell division, and in the case of DNA damage, they are key targets of the checkpoint machinery that ensure genetic stability. Little is known about the mechanisms underlying dysregulation and downstream targets of CDC25. To understand these mechanisms, we silenced the CDC25A gene in breast cancer cell line MDA-MB-231 and studied downstream targets of CDC25A gene. MDA-MB-231 breast cancer cells were transfected and silenced by CDC25A small interfering RNA. Total messenger RNA (mRNA) was extracted and analyzed by quantitative real-time polymerase chain reaction. CDC25A phosphatase level was visualized by Western blot analysis and was analyzed by 2D electrophoresis and LC-ESI-MS/MS. After CDC25A silencing, cell proliferation reduced, and the expression of 12 proteins changed. These proteins are involved in cell-cycle regulation, programmed cell death, cell differentiation, regulation of gene expression, mRNA editing, protein folding, and cell signaling pathways. Five of these proteins, including ribosomal protein lateral stalk subunit P0, growth factor receptor bound protein 2, pyruvate kinase muscle 2, eukaryotic translation elongation factor 2, and calpain small subunit 1 increase the activity of cyclin D1. Our results suggest that CDC25A controls the cell proliferation and tumorigenesis by a change in expression of proteins involved in cyclin D1 regulation and G1/S transition.
Collapse
Affiliation(s)
- Hossein Sadeghi
- Department of Genetics, Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoud Golalipour
- Department of Genetics, Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ahad Yamchi
- Department of Biotechnology, Golestan University, Gorgan, Iran
| | - Touraj Farazmandfar
- Department of Genetics, Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Department of Genetics, Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
62
|
Tossounian MA, Wahni K, Van Molle I, Vertommen D, Astolfi Rosado L, Messens J. Redox-regulated methionine oxidation of Arabidopsis thaliana glutathione transferase Phi9 induces H-site flexibility. Protein Sci 2018; 28:56-67. [PMID: 29732642 DOI: 10.1002/pro.3440] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 11/08/2022]
Abstract
Glutathione transferase enzymes help plants to cope with biotic and abiotic stress. They mainly catalyze the conjugation of glutathione (GSH) onto xenobiotics, and some act as glutathione peroxidase. With X-ray crystallography, kinetics, and thermodynamics, we studied the impact of oxidation on Arabidopsis thaliana glutathione transferase Phi 9 (GSTF9). GSTF9 has no cysteine in its sequence, and it adopts a universal GST structural fold characterized by a typical conserved GSH-binding site (G-site) and a hydrophobic co-substrate-binding site (H-site). At elevated H2 O2 concentrations, methionine sulfur oxidation decreases its transferase activity. This oxidation increases the flexibility of the H-site loop, which is reflected in lower activities for hydrophobic substrates. Determination of the transition state thermodynamic parameters shows that upon oxidation an increased enthalpic penalty is counterbalanced by a more favorable entropic contribution. All in all, to guarantee functionality under oxidative stress conditions, GSTF9 employs a thermodynamic and structural compensatory mechanism and becomes substrate of methionine sulfoxide reductases, making it a redox-regulated enzyme.
Collapse
Affiliation(s)
- Maria-Armineh Tossounian
- VIB-VUB Center for Structural Biology, Brussels, B-1050, Belgium.,Brussels Center for Redox Biology, Brussels, B-1050, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, B-1050, Belgium
| | - Khadija Wahni
- VIB-VUB Center for Structural Biology, Brussels, B-1050, Belgium.,Brussels Center for Redox Biology, Brussels, B-1050, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, B-1050, Belgium
| | - Inge Van Molle
- VIB-VUB Center for Structural Biology, Brussels, B-1050, Belgium.,Brussels Center for Redox Biology, Brussels, B-1050, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, B-1050, Belgium
| | - Didier Vertommen
- de Duve Institute, Université Catholique de Louvain, Brussels, B-1200, Belgium
| | - Leonardo Astolfi Rosado
- VIB-VUB Center for Structural Biology, Brussels, B-1050, Belgium.,Brussels Center for Redox Biology, Brussels, B-1050, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, B-1050, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Brussels, B-1050, Belgium.,Brussels Center for Redox Biology, Brussels, B-1050, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, B-1050, Belgium
| |
Collapse
|
63
|
Zhang J, Ye ZW, Singh S, Townsend DM, Tew KD. An evolving understanding of the S-glutathionylation cycle in pathways of redox regulation. Free Radic Biol Med 2018; 120:204-216. [PMID: 29578070 PMCID: PMC5940525 DOI: 10.1016/j.freeradbiomed.2018.03.038] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022]
Abstract
By nature of the reversibility of the addition of glutathione to low pKa cysteine residues, the post-translational modification of S-glutathionylation sanctions a cycle that can create a conduit for cell signaling events linked with cellular exposure to oxidative or nitrosative stress. The modification can also avert proteolysis by protection from over-oxidation of those clusters of target proteins that are substrates. Altered functions are associated with S-glutathionylation of proteins within the mitochondria and endoplasmic reticulum compartments, and these impact energy production and protein folding pathways. The existence of human polymorphisms of enzymes involved in the cycle (particularly glutathione S-transferase P) create a scenario for inter-individual variance in response to oxidative stress and a number of human diseases with associated aberrant S-glutathionylation have now been identified.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, United States
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, United States
| | - Shweta Singh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street, MSC141, Charleston, SC 29425, United States
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, United States.
| |
Collapse
|
64
|
Xie Y, Dahlin JL, Oakley AJ, Casarotto MG, Board PG, Baell JB. Reviewing Hit Discovery Literature for Difficult Targets: Glutathione Transferase Omega-1 as an Example. J Med Chem 2018; 61:7448-7470. [PMID: 29652143 DOI: 10.1021/acs.jmedchem.8b00318] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Early stage drug discovery reporting on relatively new or difficult targets is often associated with insufficient hit triage. Literature reviews of such targets seldom delve into the detail required to critically analyze the associated screening hits reported. Here we take the enzyme glutathione transferase omega-1 (GSTO1-1) as an example of a relatively difficult target and review the associated literature involving small-molecule inhibitors. As part of this process we deliberately pay closer-than-usual attention to assay interference and hit quality aspects. We believe this Perspective will be a useful guide for future development of GSTO1-1 inhibitors, as well serving as a template for future review formats of new or difficult targets.
Collapse
Affiliation(s)
- Yiyue Xie
- Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia
| | - Jayme L Dahlin
- Department of Pathology , Brigham and Women's Hospital , Boston , Massachusetts 02135 , United States
| | - Aaron J Oakley
- School of Chemistry , University of Wollongong , Wollongong , NSW 2522 , Australia
| | - Marco G Casarotto
- John Curtin School of Medical Research , Australian National University , Canberra , ACT 2600 , Australia
| | - Philip G Board
- John Curtin School of Medical Research , Australian National University , Canberra , ACT 2600 , Australia
| | - Jonathan B Baell
- Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia.,School of Pharmaceutical Sciences , Nanjing Tech University , Nanjing , 211816 , People's Republic of China
| |
Collapse
|
65
|
Foote AP, Keel BN, Zarek CM, Lindholm-Perry AK. Beef steers with average dry matter intake and divergent average daily gain have altered gene expression in the jejunum. J Anim Sci 2018; 95:4430-4439. [PMID: 29108031 DOI: 10.2527/jas2017.1804] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The objective of this study was to determine the association of differentially expressed genes (DEG) in the jejunum of steers with average DMI and high or low ADG. Feed intake and growth were measured in a cohort of 144 commercial Angus steers consuming a finishing diet containing (on a DM basis) 67.8% dry-rolled corn, 20% wet distillers grains with solubles, 8% alfalfa hay, and 4.2% vitamin/mineral supplement. From the cohort, a subset of steers with DMI within ±0.32 SD of the mean for DMI and the greatest (high) and least (low) ADG were chosen for slaughter and jejunum mucosa collection ( = 8 for each group). Dry matter intake (10.1 ± 0.05 kg/d) was not different ( = 0.41) but ADG was greater in the high-gain group (2.17 and 1.72 ± 0.02 kg/d for the high- and low-ADG groups, respectively; < 0.01). A total of 13,747 genes were found to be expressed in the jejunum, of which 64 genes were differentially expressed between the 2 groups (corrected < 0.05). Ten of the DEG were upregulated in the low-ADG group and 54 were upregulated in the high-ADG group. Gene ontology analysis determined that 24 biological process terms were overrepresented ( < 0.05), including digestion, drug and xenobiotic metabolism, and carbohydrate metabolism. Additionally, 89 molecular function terms were enriched ( < 0.05), including metallopeptidase activity, transporter activity, steroid hydrolase activity, glutathione transferase activity, and chemokine receptor binding. Metabolic pathways (28 pathways) impacted by the DEG ( < 0.05) included drug and xenobiotic metabolism by cytochrome P450, carbohydrate digestion and absorption, vitamin digestion and absorption, galactose metabolism, and linoleic acid metabolism. Results from this experiment indicate that cattle with average DMI and greater ADG likely have a greater capacity to handle foreign substances (xenobiotics). It is also possible that cattle with a greater ADG have a greater potential to digest and absorb nutrients in the small intestine.
Collapse
|
66
|
Kekulandara DN, Samarasinghe KTG, Munkanatta Godage DNP, Ahn YH. Clickable glutathione using tetrazine-alkene bioorthogonal chemistry for detecting protein glutathionylation. Org Biomol Chem 2018; 14:10886-10893. [PMID: 27812596 DOI: 10.1039/c6ob02050j] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Protein glutathionylation is one of the major cysteine oxidative modifications in response to reactive oxygen species (ROS). We recently developed a clickable glutathione approach for detecting glutathionylation by using a glutathione synthetase mutant (GS M4) that synthesizes azido-glutathione (γGlu-Cys-azido-Ala) in situ in cells. In order to demonstrate the versatility of clickable glutathione and to increase the chemical tools for detecting glutathionylation, we sought to develop clickable glutathione that uses tetrazine-alkene bioorthogonal chemistry. Here we report two alkene-containing glycine surrogates (allyl-Gly and allyl-Ser) for the biosynthesis of clickable glutathione and their use for detection, enrichment, and identification of glutathionylated proteins. Our results provide chemical tools (allyl-Gly and allyl-Ser for GS M4) for versatile characterization of protein glutathionylation. In addition, we show that the active site of GS can be tuned to introduce a small size chemical tag on glutathione for exploring glutathione function in cells.
Collapse
Affiliation(s)
| | | | | | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| |
Collapse
|
67
|
Abstract
Many potentially toxic electrophilic xenobiotics and some endogenous compounds are detoxified by conversion to the corresponding glutathione S-conjugate, which is metabolized to the N-acetylcysteine S-conjugate (mercapturate) and excreted. Some mercapturate pathway components, however, are toxic. Bioactivation (toxification) may occur when the glutathione S-conjugate (or mercapturate) is converted to a cysteine S-conjugate that undergoes a β-lyase reaction. If the sulfhydryl-containing fragment produced in this reaction is reactive, toxicity may ensue. Some drugs and halogenated workplace/environmental contaminants are bioactivated by this mechanism. On the other hand, cysteine S-conjugate β-lyases occur in nature as a means of generating some biologically useful sulfhydryl-containing compounds.
Collapse
|
68
|
Omega Class Glutathione S-Transferase: Antioxidant Enzyme in Pathogenesis of Neurodegenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5049532. [PMID: 29435097 PMCID: PMC5757135 DOI: 10.1155/2017/5049532] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/26/2017] [Indexed: 02/06/2023]
Abstract
The omega class glutathione S-transferases (GSTOs) are multifunctional enzymes involved in cellular defense and have distinct structural and functional characteristics, which differ from those of other GSTs. Previous studies provided evidence for the neuroprotective effects of GSTOs. However, the molecular mechanisms underpinning the neuroprotective functions of GSTOs have not been fully elucidated. Recently, our genetic and molecular studies using the Drosophila system have suggested that GstO1 has a protective function against H2O2-induced neurotoxicity by regulating the MAPK signaling pathway, and GstO2 is required for the activation of mitochondrial ATP synthase in the Drosophila neurodegenerative disease model. The comprehensive understanding of various neuroprotection mechanisms of Drosophila GstOs from our studies provides valuable insight into the neuroprotective functions of GstOs in vivo. In this review, we briefly introduce recent studies and summarize the novel biological functions and mechanisms underpinning neuroprotective effects of GstOs in Drosophila.
Collapse
|
69
|
Menon D, Innes A, Oakley AJ, Dahlstrom JE, Jensen LM, Brüstle A, Tummala P, Rooke M, Casarotto MG, Baell JB, Nguyen N, Xie Y, Cuellar M, Strasser J, Dahlin JL, Walters MA, Burgio G, O’Neill LAJ, Board PG. GSTO1-1 plays a pro-inflammatory role in models of inflammation, colitis and obesity. Sci Rep 2017; 7:17832. [PMID: 29259211 PMCID: PMC5736720 DOI: 10.1038/s41598-017-17861-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 12/01/2017] [Indexed: 01/07/2023] Open
Abstract
Glutathione transferase Omega 1 (GSTO1-1) is an atypical GST reported to play a pro-inflammatory role in response to LPS. Here we show that genetic knockout of Gsto1 alters the response of mice to three distinct inflammatory disease models. GSTO1-1 deficiency ameliorates the inflammatory response stimulated by LPS and attenuates the inflammatory impact of a high fat diet on glucose tolerance and insulin resistance. In contrast, GSTO1-1 deficient mice show a more severe inflammatory response and increased escape of bacteria from the colon into the lymphatic system in a dextran sodium sulfate mediated model of inflammatory bowel disease. These responses are similar to those of TLR4 and MyD88 deficient mice in these models and confirm that GSTO1-1 is critical for a TLR4-like pro-inflammatory response in vivo. In wild-type mice, we show that a small molecule inhibitor that covalently binds in the active site of GSTO1-1 can be used to ameliorate the inflammatory response to LPS. Our findings demonstrate the potential therapeutic utility of GSTO1-1 inhibitors in the modulation of inflammation and suggest their possible application in the treatment of a range of inflammatory conditions.
Collapse
Affiliation(s)
- Deepthi Menon
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia ,0000 0004 1936 9705grid.8217.cSchool of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Ashlee Innes
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Aaron J. Oakley
- 0000 0004 0486 528Xgrid.1007.6School of Chemistry, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Jane E. Dahlstrom
- 0000 0000 9984 5644grid.413314.0ACT Pathology and ANU Medical School, The Canberra Hospital, Garran, ACT 2605 Australia
| | - Lora M. Jensen
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Anne Brüstle
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Padmaja Tummala
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Melissa Rooke
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Marco G. Casarotto
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Jonathan B. Baell
- 0000 0004 1936 7857grid.1002.3Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia ,0000 0000 9389 5210grid.412022.7School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 People’s Republic of China
| | - Nghi Nguyen
- 0000 0004 1936 7857grid.1002.3Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Yiyue Xie
- 0000 0004 1936 7857grid.1002.3Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Matthew Cuellar
- 0000000419368657grid.17635.36Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN USA
| | - Jessica Strasser
- 0000000419368657grid.17635.36Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN USA
| | - Jayme L. Dahlin
- 0000 0004 0378 8294grid.62560.37Department of Pathology, Brigham and Women’s Hospital, Boston, MA USA
| | - Michael A. Walters
- 0000000419368657grid.17635.36Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN USA
| | - Gaetan Burgio
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| | - Luke A. J. O’Neill
- 0000 0004 1936 9705grid.8217.cSchool of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Philip G. Board
- 0000 0001 2180 7477grid.1001.0John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600 Australia
| |
Collapse
|
70
|
Regulation of protein function by S-nitrosation and S-glutathionylation: processes and targets in cardiovascular pathophysiology. Biol Chem 2017; 398:1267-1293. [DOI: 10.1515/hsz-2017-0150] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023]
Abstract
AbstractDecades of chemical, biochemical and pathophysiological research have established the relevance of post-translational protein modifications induced by processes related to oxidative stress, with critical reflections on cellular signal transduction pathways. A great deal of the so-called ‘redox regulation’ of cell function is in fact mediated through reactions promoted by reactive oxygen and nitrogen species on more or less specific aminoacid residues in proteins, at various levels within the cell machinery. Modifications involving cysteine residues have received most attention, due to the critical roles they play in determining the structure/function correlates in proteins. The peculiar reactivity of these residues results in two major classes of modifications, with incorporation of NO moieties (S-nitrosation, leading to formation of proteinS-nitrosothiols) or binding of low molecular weight thiols (S-thionylation, i.e. in particularS-glutathionylation,S-cysteinylglycinylation andS-cysteinylation). A wide array of proteins have been thus analyzed in detail as far as their susceptibility to either modification or both, and the resulting functional changes have been described in a number of experimental settings. The present review aims to provide an update of available knowledge in the field, with a special focus on the respective (sometimes competing and antagonistic) roles played by proteinS-nitrosations andS-thionylations in biochemical and cellular processes specifically pertaining to pathogenesis of cardiovascular diseases.
Collapse
|
71
|
Solution structure of the TLR adaptor MAL/TIRAP reveals an intact BB loop and supports MAL Cys91 glutathionylation for signaling. Proc Natl Acad Sci U S A 2017; 114:E6480-E6489. [PMID: 28739909 DOI: 10.1073/pnas.1701868114] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MyD88 adaptor-like (MAL) is a critical protein in innate immunity, involved in signaling by several Toll-like receptors (TLRs), key pattern recognition receptors (PRRs). Crystal structures of MAL revealed a nontypical Toll/interleukin-1 receptor (TIR)-domain fold stabilized by two disulfide bridges. We therefore undertook a structural and functional analysis of the role of reactive cysteine residues in the protein. Under reducing conditions, the cysteines do not form disulfides, but under oxidizing conditions they are highly amenable to modification. The solution structure of the reduced form of the MAL TIR domain, determined by NMR spectroscopy, reveals a remarkable structural rearrangement compared with the disulfide-bonded structure, which includes the relocation of a β-strand and repositioning of the functionally important "BB-loop" region to a location more typical for TIR domains. Redox measurements by NMR further reveal that C91 has the highest redox potential of all cysteines in MAL. Indeed, mass spectrometry revealed that C91 undergoes glutathionylation in macrophages activated with the TLR4 ligand lipopolysaccharide (LPS). The C91A mutation limits MAL glutathionylation and acts as a dominant negative, blocking the interaction of MAL with its downstream target MyD88. The H92P mutation mimics the dominant-negative effects of the C91A mutation, presumably by preventing C91 glutathionylation. The MAL C91A and H92P mutants also display diminished degradation and interaction with interleukin-1 receptor-associated kinase 4 (IRAK4). We conclude that in the cell, MAL is not disulfide-bonded and requires glutathionylation of C91 for signaling.
Collapse
|
72
|
Glutathione and Glutathione Transferase Omega 1 as Key Posttranslational Regulators in Macrophages. Microbiol Spectr 2017; 5. [PMID: 28102119 DOI: 10.1128/microbiolspec.mchd-0044-2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophage activation during phagocytosis or by pattern recognition receptors, such as Toll-like receptor 4, leads to the accumulation of reactive oxygen species (ROS). ROS act as a microbicidal defense mechanism, promoting clearance of infection, allowing for resolution of inflammation. Overproduction of ROS, however, overwhelms our cellular antioxidant defense system, promoting oxidation of protein machinery, leading to macrophage dysregulation and pathophysiology of chronic inflammatory conditions, such as atherosclerosis. Here we will describe the role of the antioxidant tripeptide glutathione (GSH). Until recently, the binding of GSH, termed glutathionylation, was only considered to maintain the integrity of cellular components, limiting the damaging effects of an aberrant oxidative environment. GSH can, however, have positive and negative regulatory effects on protein function in macrophages. GSH regulates protein secretion, driving tumor necrosis factor α release, hypoxia-inducible factor-1α stability, STAT3 phosphorylation, and caspase-1 activation in macrophages. GSH also plays a role in host defense against Listeria monocytogenes, modifying the key virulence protein PrfA in infected macrophages. We will also discuss glutathione transferase omega 1, a deglutathionylating enzyme recently shown to play a role in many aspects of macrophage activity, including metabolism, NF-κB activation, and cell survival pathways. Glutathionylation is emerging as a key regulatory event in macrophage biology that might be susceptible to therapeutic targeting.
Collapse
|
73
|
Djukic T, Simic T, Pljesa-Ercegovac M, Matic M, Suvakov S, Coric V, Dragicevic D, Savic-Radojevic A. Upregulated glutathione transferase omega-1 correlates with progression of urinary bladder carcinoma. Redox Rep 2017; 22:486-492. [PMID: 28288548 DOI: 10.1080/13510002.2017.1299909] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVES Newly discovered glutathione transferase omega 1 (GSTO1-1) plays an important role in the glutathionylation cycle, a significant mechanism of protein function regulation. GSTO1-1 expression pattern has not been studied in transitional cell carcinoma (TCC), as yet. METHODS A total of 56 TCC tumor and corresponding non-tumor specimens were investigated. Glutathione content and thioltransferase activity were measured spectrophotometrically. Protein-glutathione mixed disulfides were measured fluorimetrically. GSTO1-1 expression was determined by immunoblot and qPCR. Immunoprecipitation with GSTO1-1 antibody was followed by immunoblot using anti-GSTO1, GSTP1, c-Jun, JNK, Akt, phospho-Akt, and ASK1 antibody, while for the total S-glutathionylation levels non-reducing electrophoresis was performed. RESULTS The contents of reduced glutathione and thioltransferase activity were significantly increased in tumor compared to non-tumor tissue. The increased GSTO1 expression in tumor tissue showed clear correlation with grade and stage. However, decreased total protein glutathionylation level in tumor compared to non-tumor samples was found. Immunoprecipitation has shown an association of GSTO1-1 with GSTP1, Akt, phospho-Akt, and ASK1 proteins. CONCLUSIONS GSTO1 deglutathionylase activity suggests its potential important role in redox perturbations present in TCC. Increased GSTO1-1 expression might contribute to TCC development and/or progression supporting the notion that GSTO1-1 may be a promising novel cancer target.
Collapse
Affiliation(s)
- Tatjana Djukic
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Tatjana Simic
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Marija Pljesa-Ercegovac
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Marija Matic
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Sonja Suvakov
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Vesna Coric
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Dejan Dragicevic
- b Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Ana Savic-Radojevic
- a Institute of Medical and Clinical Biochemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| |
Collapse
|
74
|
Advances in drug metabolism and pharmacogenetics research in Australia. Pharmacol Res 2017; 116:7-19. [DOI: 10.1016/j.phrs.2016.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 01/04/2023]
|
75
|
Chandel A, Bachhawat AK. Redox regulation of the yeast voltage-gated calcium channel homolog, Cch1p, by glutathionylation of specific cysteines. J Cell Sci 2017; 130:2317-2328. [DOI: 10.1242/jcs.202853] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/26/2017] [Indexed: 01/12/2023] Open
Abstract
CCH1, the yeast homolog of the pore-forming subunit α1 of the mammalian Voltage-gated calcium channel (VGCC) located on the plasma membrane mediates the redox-dependent influx of calcium. Cch1p is known to undergo both rapid activation (oxidative stress, high pH) and slow activation (ER stress, mating pheromone activation), but the mechanism of activation is not known. We demonstrate here that the fast activation, as well as the slow activation (tunicamycin or α-factor) is mediated through a common redox-dependant manner. Further, through mutational analysis of all 18 exposed cysteines in the Cch1p protein, we show that four of these mutants, C587A, C606A, C636A and C642A, which are clustered together in a common cytoplasmic loop region were functionally defective during both fast and slow activations and also showed reduced glutathionylation. These four cysteines are also conserved across phyla suggesting a conserved mechanism of activation. Investigations into the enzymes involved in the activation reveal that the yeast glutathione-s-transferase, Gtt1p is involved in the glutathionylation of Cch1p, while the thioredoxin, Trx2p plays a role in the Cch1p deglutathionylation.
Collapse
Affiliation(s)
- Avinash Chandel
- Department of Biological Sciences, Indian Institute of Science Education & Research (IISER), Sector 81, Mohali, 140306, Punjab, India
| | - Anand K. Bachhawat
- Department of Biological Sciences, Indian Institute of Science Education & Research (IISER), Sector 81, Mohali, 140306, Punjab, India
| |
Collapse
|
76
|
Short JD, Downs K, Tavakoli S, Asmis R. Protein Thiol Redox Signaling in Monocytes and Macrophages. Antioxid Redox Signal 2016; 25:816-835. [PMID: 27288099 PMCID: PMC5107717 DOI: 10.1089/ars.2016.6697] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Monocyte and macrophage dysfunction plays a critical role in a wide range of inflammatory disease processes, including obesity, impaired wound healing diabetic complications, and atherosclerosis. Emerging evidence suggests that the earliest events in monocyte or macrophage dysregulation include elevated reactive oxygen species production, thiol modifications, and disruption of redox-sensitive signaling pathways. This review focuses on the current state of research in thiol redox signaling in monocytes and macrophages, including (i) the molecular mechanisms by which reversible protein-S-glutathionylation occurs, (ii) the identification of bona fide S-glutathionylated proteins that occur under physiological conditions, and (iii) how disruptions of thiol redox signaling affect monocyte and macrophage functions and contribute to atherosclerosis. Recent Advances: Recent advances in redox biochemistry and biology as well as redox proteomic techniques have led to the identification of many new thiol redox-regulated proteins and pathways. In addition, major advances have been made in expanding the list of S-glutathionylated proteins and assessing the role that protein-S-glutathionylation and S-glutathionylation-regulating enzymes play in monocyte and macrophage functions, including monocyte transmigration, macrophage polarization, foam cell formation, and macrophage cell death. CRITICAL ISSUES Protein-S-glutathionylation/deglutathionylation in monocytes and macrophages has emerged as a new and important signaling paradigm, which provides a molecular basis for the well-established relationship between metabolic disorders, oxidative stress, and cardiovascular diseases. FUTURE DIRECTIONS The identification of specific S-glutathionylated proteins as well as the mechanisms that control this post-translational protein modification in monocytes and macrophages will facilitate the development of new preventive and therapeutic strategies to combat atherosclerosis and other metabolic diseases. Antioxid. Redox Signal. 25, 816-835.
Collapse
Affiliation(s)
- John D Short
- 1 Department of Pharmacology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Kevin Downs
- 2 Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Sina Tavakoli
- 3 Department of Radiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Reto Asmis
- 4 Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,5 Department of Biochemistry, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| |
Collapse
|
77
|
Mechanistic evaluation and transcriptional signature of a glutathione S-transferase omega 1 inhibitor. Nat Commun 2016; 7:13084. [PMID: 27703239 PMCID: PMC5059489 DOI: 10.1038/ncomms13084] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/01/2016] [Indexed: 12/26/2022] Open
Abstract
Glutathione S-transferase omega 1 (GSTO1) is an atypical GST isoform that is overexpressed in several cancers and has been implicated in drug resistance. Currently, no small-molecule drug targeting GSTO1 is under clinical development. Here we show that silencing of GSTO1 with siRNA significantly impairs cancer cell viability, validating GSTO1 as a potential new target in oncology. We report on the development and characterization of a series of chloroacetamide-containing potent GSTO1 inhibitors. Co-crystal structures of GSTO1 with our inhibitors demonstrate covalent binding to the active site cysteine. These potent GSTO1 inhibitors suppress cancer cell growth, enhance the cytotoxic effects of cisplatin and inhibit tumour growth in colon cancer models as single agent. Bru-seq-based transcription profiling unravelled novel roles for GSTO1 in cholesterol metabolism, oxidative and endoplasmic stress responses, cytoskeleton and cell migration. Our findings demonstrate the therapeutic utility of GSTO1 inhibitors as anticancer agents and identify the novel cellular pathways under GSTO1 regulation in colorectal cancer. Glutathione S-transferase omega 1 (GSTO1) is an atypical GST isoform overexpressed in several cancers that has been implicated in drug resistance. Here the authors identify a small molecule inhibitor of GSTO1 that effectively inhibits tumor growth in colon cancer models, and establish its mechanism of action.
Collapse
|
78
|
Chandel A, Das KK, Bachhawat AK. Glutathione depletion activates the yeast vacuolar transient receptor potential channel, Yvc1p, by reversible glutathionylation of specific cysteines. Mol Biol Cell 2016; 27:3913-3925. [PMID: 27708136 PMCID: PMC5170613 DOI: 10.1091/mbc.e16-05-0281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/14/2016] [Accepted: 09/27/2016] [Indexed: 11/12/2022] Open
Abstract
Glutathione depletion leads to calcium influx in yeast cells via plasma membrane Cch1p and the vacuolar Yvc1p channels. Yvc1p, a yeast vacuolar transient receptor potential channel, is activated by glutathionylation carried out by the glutathione S-transferase Gtt1p, and this mechanism is reversible with deglutathionylation being mediated by the thioredoxin Trx2p. Glutathione depletion and calcium influx into the cytoplasm are two hallmarks of apoptosis. We have been investigating how glutathione depletion leads to apoptosis in yeast. We show here that glutathione depletion in yeast leads to the activation of two cytoplasmically inward-facing channels: the plasma membrane, Cch1p, and the vacuolar calcium channel, Yvc1p. Deletion of these channels partially rescues cells from glutathione depletion–induced cell death. Subsequent investigations on the Yvc1p channel, a homologue of the mammalian TRP channels, revealed that the channel is activated by glutathionylation. Yvc1p has nine cysteine residues, of which eight are located in the cytoplasmic regions and one on the transmembrane domain. We show that three of these cysteines, Cys-17, Cys-79, and Cys-191, are specifically glutathionylated. Mutation of these cysteines to alanine leads to a loss in glutathionylation and a concomitant loss in calcium channel activity. We further investigated the mechanism of glutathionylation and demonstrate a role for the yeast glutathione S-transferase Gtt1p in glutathionylation. Yvc1p is also deglutathionylated, and this was found to be mediated by the yeast thioredoxin, Trx2p. A model for redox activation and deactivation of the yeast Yvc1p channel is presented.
Collapse
Affiliation(s)
- Avinash Chandel
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| | - Krishna K Das
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| | - Anand K Bachhawat
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| |
Collapse
|
79
|
Jiang P, Vegge A, Thymann T, Wan JMF, Sangild PT. Glucagon-Like Peptide 2 Stimulates Postresection Intestinal Adaptation in Preterm Pigs by Affecting Proteins Related to Protein, Carbohydrate, and Sulphur Metabolism. JPEN J Parenter Enteral Nutr 2016; 41:1293-1300. [DOI: 10.1177/0148607116662971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Pingping Jiang
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Andreas Vegge
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Global Research, Novo Nordisk, Måløv, Denmark
| | - Thomas Thymann
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Per Torp Sangild
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
80
|
Bogdanova A, Petrushanko IY, Hernansanz-Agustín P, Martínez-Ruiz A. "Oxygen Sensing" by Na,K-ATPase: These Miraculous Thiols. Front Physiol 2016; 7:314. [PMID: 27531981 PMCID: PMC4970491 DOI: 10.3389/fphys.2016.00314] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/12/2016] [Indexed: 12/16/2022] Open
Abstract
Control over the Na,K-ATPase function plays a central role in adaptation of the organisms to hypoxic and anoxic conditions. As the enzyme itself does not possess O2 binding sites its "oxygen-sensitivity" is mediated by a variety of redox-sensitive modifications including S-glutathionylation, S-nitrosylation, and redox-sensitive phosphorylation. This is an overview of the current knowledge on the plethora of molecular mechanisms tuning the activity of the ATP-consuming Na,K-ATPase to the cellular metabolic activity. Recent findings suggest that oxygen-derived free radicals and H2O2, NO, and oxidized glutathione are the signaling messengers that make the Na,K-ATPase "oxygen-sensitive." This very ancient signaling pathway targeting thiols of all three subunits of the Na,K-ATPase as well as redox-sensitive kinases sustains the enzyme activity at the "optimal" level avoiding terminal ATP depletion and maintaining the transmembrane ion gradients in cells of anoxia-tolerant species. We acknowledge the complexity of the underlying processes as we characterize the sources of reactive oxygen and nitrogen species production in hypoxic cells, and identify their targets, the reactive thiol groups which, upon modification, impact the enzyme activity. Structured accordingly, this review presents a summary on (i) the sources of free radical production in hypoxic cells, (ii) localization of regulatory thiols within the Na,K-ATPase and the role reversible thiol modifications play in responses of the enzyme to a variety of stimuli (hypoxia, receptors' activation) (iii) redox-sensitive regulatory phosphorylation, and (iv) the role of fine modulation of the Na,K-ATPase function in survival success under hypoxic conditions. The co-authors attempted to cover all the contradictions and standing hypotheses in the field and propose the possible future developments in this dynamic area of research, the importance of which is hard to overestimate. Better understanding of the processes underlying successful adaptation strategies will make it possible to harness them and use for treatment of patients with stroke and myocardial infarction, sleep apnoea and high altitude pulmonary oedema, and those undergoing surgical interventions associated with the interruption of blood perfusion.
Collapse
Affiliation(s)
- Anna Bogdanova
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of ZurichZurich, Switzerland
| | - Irina Y. Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of SciencesMoscow, Russia
| | - Pablo Hernansanz-Agustín
- Servicio de Inmunología, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La PrincesaMadrid, Spain
- Departamento de Bioquímica, Universidad Autónoma de MadridMadrid, Spain
| | - Antonio Martínez-Ruiz
- Servicio de Inmunología, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La PrincesaMadrid, Spain
| |
Collapse
|
81
|
Samarasinghe KTG, Munkanatta Godage DNP, Zhou Y, Ndombera FT, Weerapana E, Ahn YH. A clickable glutathione approach for identification of protein glutathionylation in response to glucose metabolism. MOLECULAR BIOSYSTEMS 2016; 12:2471-80. [PMID: 27216279 PMCID: PMC4955733 DOI: 10.1039/c6mb00175k] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glucose metabolism and mitochondrial function are closely interconnected with cellular redox-homeostasis. Although glucose starvation, which mimics ischemic conditions or insufficient vascularization, is known to perturb redox-homeostasis, global and individual protein glutathionylation in response to glucose metabolism or mitochondrial activity remains largely unknown. In this report, we use our clickable glutathione approach, which forms clickable glutathione (azido-glutathione) by using a mutant of glutathione synthetase (GS M4), for detection and identification of protein glutathionylation in response to glucose starvation. We found that protein glutathionylation is readily induced in HEK293 cells in response to low glucose concentrations when mitochondrial reactive oxygen species (ROS) are elevated in cells, and glucose is the major determinant for inducing reversible glutathionylation. Proteomic and biochemical analysis identified over 1300 proteins, including SMYD2, PP2Cα, and catalase. We further showed that PP2Cα is glutathionylated at C314 in a C-terminal domain, and PP2Cα C314 glutathionylation disrupts the interaction with mGluR3, an important glutamate receptor associated with synaptic plasticity.
Collapse
Affiliation(s)
| | | | - Yani Zhou
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | - Fidelis T Ndombera
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| | - Eranthie Weerapana
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA.
| |
Collapse
|
82
|
Cimbaljevic S, Suvakov S, Matic M, Pljesa-Ercegovac M, Pekmezovic T, Radic T, Coric V, Damjanovic T, Dimkovic N, Markovic R, Savic-Radojevic A, Simic T. Association of GSTO1 and GSTO2 Polymorphism with Risk of End-Stage Renal Disease Development and Patient Survival. J Med Biochem 2016; 35:302-311. [PMID: 28356881 PMCID: PMC5346808 DOI: 10.1515/jomb-2016-0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/12/2016] [Indexed: 11/19/2022] Open
Abstract
Background Oxidative stress in patients with end-stage renal disease (ESRD) is associated with long-term cardiovascular complications. The cytosolic family of glutathione S-transferases (GSTs) is involved in the detoxication of various toxic compounds and antioxidant protection. GST omega class members, GSTO1 and GSTO2 possess, unlike other GSTs, dehydroascorbate reductase and deglutathionylation activities. The aim of this study was to clarify the role of genetic polymorphisms of GSTO1 (rs4925) and GSTO2 (rs156697) as risk determinants for ESRD development, as well as in the survival of these patients. Methods A total of 199 patients and 199 healthy subjects were included in the study and genotyped for both GSTO1 and GSTO2 polymorphism. Protein thiol and carbonyl groups as markers of protein oxidative damage were determined spectrophotometrically. Cox proportional hazard model and Kaplan-Meier analysis were performed to investigate the role of GSTO1 and GSTO2 genetic polymorphism on mortality of ESRD patients during the follow-up period (36 month). Results Individuals carrying the variant GSTO2 GG genotype were at 2.45-fold higher risk of ESRD development compared to the wild type GSTO2 AA genotype (OR=2.45; 95%CI=1.18–5.07; p=0.016). The results of GSTO1/GSTO2 haplotype analysis showed that the haplotype combination of GSTO1 (*A)/GSTO2 (*A) (GSTO1 variant/GSTO2 wild type allele) was protective for ESRD (OR=0.23 95%CI=0.12-0.44, p=0.001). Patients carrying at least one GSTO1 reference allele have shorter mean overall (Log rank=2.844, p =0.241) and cardiovascular survival probability (Log rank=4.211, p=0.122). Conclusions GSTO polymorphisms have been shown to act as significant markers in assessing the risk of ESRD development and patients’ survival.
Collapse
Affiliation(s)
| | - Sonja Suvakov
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana Pekmezovic
- Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tanja Radic
- 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
| | - Tatjana Damjanovic
- Clinical Department for Renal Diseases, Zvezdara University Medical Center, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nada Dimkovic
- Clinical Department for Renal Diseases, Zvezdara University Medical Center, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Rodoljub Markovic
- Department of Nephrology and Hemodialysis, University Teaching Hospital Zemun, Belgrade, Serbia
| | - Ana Savic-Radojevic
- 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
|
83
|
Kim JG, Ahn CS, Kim SH, Bae YA, Kwon NY, Kang I, Yang HJ, Sohn WM, Kong Y. Clonorchis sinensis omega-class glutathione transferases play major roles in the protection of the reproductive system during maturation and the response to oxidative stress. Parasit Vectors 2016; 9:337. [PMID: 27296469 PMCID: PMC4906895 DOI: 10.1186/s13071-016-1622-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/02/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clonorchis sinensis causes a major food-borne helminthic infection. This species locates in mammalian hepatobiliary ducts, where oxidative stressors and hydrophobic substances are profuse. To adapt to the hostile micromilieu and to ensure its long-term survival, the parasite continuously produces a diverse repertoire of antioxidant enzymes including several species of glutathione transferases (GSTs). Helminth GSTs play pertinent roles during sequestration of harmful xenobiotics since most helminths lack the cytochrome P-450 detoxifying enzyme. METHODS We isolated and analyzed the biochemical properties of two omega-class GSTs of C. sinensis (CsGSTo1 and CsGSTo2). We observed spatiotemporal expression patterns in accordance with the maturation of the worm's reproductive system. Possible biological protective roles of CsGSTos in these organs under oxidative stress were investigated. RESULTS The full-length cDNAs of CsGSTo1 and 2 constituted 965 bp and 1,061 bp with open reading frames of 737 bp (246 amino acids) and 669 bp (223 amino acids). They harbored characteristic N-terminal thioredoxin-like and C-terminal α-helical domains. A cysteine residue, which constituted omega-class specific active site, and the glutathione-binding amino acids, were recognized in appropriate positions. They shared 44 % sequence identity with each other and 14.8-44.8 % with orthologues/homologues from other organisms. Bacterially expressed recombinant proteins (rCsGSTo1 and 2) exhibited dehydroascorbate reductase (DHAR) and thioltransferase activities. DHAR activity was higher than thioltransferase activity. They showed weak canonical GST activity toward 1-chloro-2,4-dinitrobenzene. S-hexylglutathione potently and competitively inhibited the active-site at nanomolar concentrations (0.63 and 0.58 nM for rCsGSTo1 and 2). Interestingly, rCsGSTos exhibited high enzyme activity toward mu- and theta-class GST specific substrate, 4-nitrobenzyl chloride. Expression of CsGSTo transcripts and proteins increased beginning in 2-week-old juveniles and reached their highest levels in 4-week-old adults. The proteins were mainly expressed in the elements of the reproductive system, such as vitelline follicles, testes, seminal receptacle, sperm and eggs. Oxidative stressors induced upregulated expression of CsGSTos in these organs. Regardless of oxidative stresses, CsGSTos continued to be highly expressed in eggs. CsGSTo1 or 2 overexpressing bacteria demonstrated high resistance under oxidative killing. CONCLUSIONS CsGSTos might be critically involved in protection of the reproductive system during maturation of C. sinensis worms and in response to oxidative conditions, thereby contributing to maintenance of parasite fecundity.
Collapse
Affiliation(s)
- Jeong-Geun Kim
- Department of Molecular Parasitology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Korea
| | - Chun-Seob Ahn
- Department of Molecular Parasitology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Korea
| | - Seon-Hee Kim
- Department of Microbiology, Graduate School of Medicine, Gachon University, Incheon, Korea
| | - Young-An Bae
- Department of Microbiology, Graduate School of Medicine, Gachon University, Incheon, Korea
| | - Na-Young Kwon
- Department of Molecular Parasitology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Korea
| | - Insug Kang
- Department of Molecular Biology and Biochemistry, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun-Jong Yang
- Department of Parasitology, Ewha Womans University, School of Medicine, Seoul, Korea
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Yoon Kong
- Department of Molecular Parasitology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Korea.
| |
Collapse
|
84
|
Selenophosphate synthetase 1 is an essential protein with roles in regulation of redox homoeostasis in mammals. Biochem J 2016; 473:2141-54. [PMID: 27208177 DOI: 10.1042/bcj20160393] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/16/2016] [Indexed: 12/23/2022]
Abstract
Selenophosphate synthetase (SPS) was initially detected in bacteria and was shown to synthesize selenophosphate, the active selenium donor. However, mammals have two SPS paralogues, which are designated SPS1 and SPS2. Although it is known that SPS2 catalyses the synthesis of selenophosphate, the function of SPS1 remains largely unclear. To examine the role of SPS1 in mammals, we generated a Sps1-knockout mouse and found that systemic SPS1 deficiency led to embryos that were clearly underdeveloped by embryonic day (E)8.5 and virtually resorbed by E14.5. The knockout of Sps1 in the liver preserved viability, but significantly affected the expression of a large number of mRNAs involved in cancer, embryonic development and the glutathione system. Particularly notable was the extreme deficiency of glutaredoxin 1 (GLRX1) and glutathione transferase Omega 1 (GSTO1). To assess these phenotypes at the cellular level, we targeted the removal of SPS1 in F9 cells, a mouse embryonal carcinoma (EC) cell line, which affected the glutathione system proteins and accordingly led to the accumulation of hydrogen peroxide in the cell. Furthermore, we found that several malignant characteristics of SPS1-deficient F9 cells were reversed, suggesting that SPS1 played a role in supporting and/or sustaining cancer. In addition, the overexpression of mouse or human GLRX1 led to a reversal of observed increases in reactive oxygen species (ROS) in the F9 SPS1/GLRX1-deficient cells and resulted in levels that were similar to those in F9 SPS1-sufficient cells. The results suggested that SPS1 is an essential mammalian enzyme with roles in regulating redox homoeostasis and controlling cell growth.
Collapse
|
85
|
Genomic instability and cellular stress in organ biopsies and peripheral blood lymphocytes from patients with colorectal cancer and predisposing pathologies. Oncotarget 2016; 6:14852-64. [PMID: 26046795 PMCID: PMC4558120 DOI: 10.18632/oncotarget.4032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/25/2022] Open
Abstract
Inflammatory bowel disease (IBD) and polyps, are common colorectal pathologies in western society and are risk factors for development of colorectal cancer (CRC). Genomic instability is a cancer hallmark and is connected to changes in chromosomal structure, often caused by double strand break formation (DSB), and aneuploidy. Cellular stress, may contribute to genomic instability. In colorectal biopsies and peripheral blood lymphocytes of patients with IBD, polyps and CRC, we evaluated 1) genomic instability using the γH2AX assay as marker of DSB and micronuclei in mononuclear lymphocytes kept under cytodieresis inhibition, and 2) cellular stress through expression and cellular localization of glutathione-S-transferase omega 1 (GSTO1). Colon biopsies showed γH2AX increase starting from polyps, while lymphocytes already from IBD. Micronuclei frequency began to rise in lymphocytes of subjects with polyps, suggesting a systemic genomic instability condition. Colorectal tissues lost GSTO1 expression but increased nuclear localization with pathology progression. Lymphocytes did not change GSTO1 expression and localization until CRC formation, where enzyme expression was increased. We propose that the growing genomic instability found in our patients is connected with the alteration of cellular environment. Evaluation of genomic damage and cellular stress in colorectal pathologies may facilitate prevention and management of CRC.
Collapse
|
86
|
Jones JT, Qian X, van der Velden JLJ, Chia SB, McMillan DH, Flemer S, Hoffman SM, Lahue KG, Schneider RW, Nolin JD, Anathy V, van der Vliet A, Townsend DM, Tew KD, Janssen-Heininger YMW. Glutathione S-transferase pi modulates NF-κB activation and pro-inflammatory responses in lung epithelial cells. Redox Biol 2016; 8:375-82. [PMID: 27058114 PMCID: PMC4827796 DOI: 10.1016/j.redox.2016.03.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/22/2016] [Indexed: 01/30/2023] Open
Abstract
Nuclear Factor kappa B (NF-κB) is a transcription factor family critical in the activation of pro- inflammatory responses. The NF-κB pathway is regulated by oxidant-induced post-translational modifications. Protein S-glutathionylation, or the conjugation of the antioxidant molecule, glutathione to reactive cysteines inhibits the activity of inhibitory kappa B kinase beta (IKKβ), among other NF-κB proteins. Glutathione S-transferase Pi (GSTP) is an enzyme that has been shown to catalyze protein S-glutathionylation (PSSG) under conditions of oxidative stress. The objective of the present study was to determine whether GSTP regulates NF-κB signaling, S-glutathionylation of IKK, and subsequent pro-inflammatory signaling. We demonstrated that, in unstimulated cells, GSTP associated with the inhibitor of NF-κB, IκBα. However, exposure to LPS resulted in a rapid loss of association between IκBα and GSTP, and instead led to a protracted association between IKKβ and GSTP. LPS exposure also led to increases in the S-glutathionylation of IKKβ. SiRNA-mediated knockdown of GSTP decreased IKKβ-SSG, and enhanced NF-κB nuclear translocation, transcriptional activity, and pro-inflammatory cytokine production in response to lipopolysaccharide (LPS). TLK117, an isotype-selective inhibitor of GSTP, also enhanced LPS-induced NF-κB transcriptional activity and pro-inflammatory cytokine production, suggesting that the catalytic activity of GSTP is important in repressing NF-κB activation. Expression of both wild-type and catalytically-inactive Y7F mutant GSTP significantly attenuated LPS- or IKKβ-induced production of GM-CSF. These studies indicate a complex role for GSTP in modulating NF-κB, which may involve S-glutathionylation of IKK proteins, and interaction with NF-κB family members. Our findings suggest that targeting GSTP is a potential avenue for regulating the activity of this prominent pro-inflammatory and immunomodulatory transcription factor.
Collapse
Affiliation(s)
- Jane T Jones
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Shi Biao Chia
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Stevenson Flemer
- Department of Chemistry, The University of Vermont, Burlington, VT, United States
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Robert W Schneider
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | | |
Collapse
|
87
|
Structure, function and disease relevance of Omega-class glutathione transferases. Arch Toxicol 2016; 90:1049-67. [PMID: 26993125 DOI: 10.1007/s00204-016-1691-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/10/2016] [Indexed: 12/13/2022]
Abstract
The Omega-class cytosolic glutathione transferases (GSTs) have distinct structural and functional attributes that allow them to perform novel roles unrelated to the functions of other GSTs. Mammalian GSTO1-1 has been found to play a previously unappreciated role in the glutathionylation cycle that is emerging as significant mechanism regulating protein function. GSTO1-1-catalyzed glutathionylation or deglutathionylation of a key signaling protein may explain the requirement for catalytically active GSTO1-1 in LPS-stimulated pro-inflammatory signaling through the TLR4 receptor. The observation that ML175 a specific GSTO1-1 inhibitor can block LPS-stimulated inflammatory signaling has opened a new avenue for the development of novel anti-inflammatory drugs that could be useful in the treatment of toxic shock and other inflammatory disorders. The role of GSTO2-2 remains unclear. As a dehydroascorbate reductase, it could contribute to the maintenance of cellular redox balance and it is interesting to note that the GSTO2 N142D polymorphism has been associated with multiple diseases including Alzheimer's disease, Parkinson's disease, familial amyotrophic lateral sclerosis, chronic obstructive pulmonary disease, age-related cataract and breast cancer.
Collapse
|
88
|
Paul S, Jakhar R, Bhardwaj M, Kang SC. Glutathione-S-transferase omega 1 (GSTO1-1) acts as mediator of signaling pathways involved in aflatoxin B1-induced apoptosis-autophagy crosstalk in macrophages. Free Radic Biol Med 2015; 89:1218-30. [PMID: 26561775 DOI: 10.1016/j.freeradbiomed.2015.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/19/2015] [Accepted: 11/04/2015] [Indexed: 12/29/2022]
Abstract
Aflatoxin B1 (AFB1) is the most toxic aflatoxin species and has been shown to be associated with specific as well as non-specific immune responses. In the present study, using murine macrophage Raw 264.7 cells as a model, we report that short exposure (6h) to AFB1 caused an increase in the cellular calcium pool in mitochondria, which in turn elevated reactive oxygen species (ROS)-mediated oxidative stress and led to loss of mitochondrial membrane potential and ultimately c-Jun N-terminal kinases (JNK)-mediated caspase-dependent cell death. On the contrary, longer exposure (12h) to AFB1 reduced JNK phosphorylation and cell death in macrophages. Measurement of autophagic flux demonstrated that autophagy induction through the canonical pathway was responsible for suppressing AFB1-induced apoptosis after 12h. As a detailed molecular mechanism, we found that the unfolded protein response (UPR) machinery was active at 12h post-exposure to AFB1 and induced cytoprotective autophagy as confirmed by determination of major autophagic markers. Inhibition of autophagy by Beclin-1 siRNA also resulted in JNK-mediated cell death. We further established that glutathione S transferase omega1-1 (GSTO1-1), a specific class of GST, was the responsible factor between apoptosis and autophagy crosstalk. Targeting of GSTO1-1 increased JNK-mediated apoptosis by 2-fold compared to the control, whereas autophagy rate was reduced. Thus, increased expression of GSTO1-1 was associated with increased protein glutathionylation, an important protein modification in response to cellular redox status.
Collapse
Affiliation(s)
- Souren Paul
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Rekha Jakhar
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Monika Bhardwaj
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Kyoungsan, Kyoungbook 712-714, Republic of Korea.
| |
Collapse
|
89
|
Németi B, Poór M, Gregus Z. Reduction of the Pentavalent Arsenical Dimethylarsinic Acid and the GSTO1 Substrate S-(4-Nitrophenacyl)glutathione by Rat Liver Cytosol: Analyzing the Role of GSTO1 in Arsenic Reduction. Chem Res Toxicol 2015; 28:2199-209. [DOI: 10.1021/acs.chemrestox.5b00368] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Balázs Németi
- Department of Pharmacology
and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, Pécs, Hungary
| | - Miklós Poór
- Department of Pharmacology
and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, Pécs, Hungary
| | - Zoltán Gregus
- Department of Pharmacology
and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, Pécs, Hungary
| |
Collapse
|
90
|
McGarry DJ, Chakravarty P, Wolf CR, Henderson CJ. Altered protein S-glutathionylation identifies a potential mechanism of resistance to acetaminophen-induced hepatotoxicity. J Pharmacol Exp Ther 2015; 355:137-44. [PMID: 26311813 PMCID: PMC4631951 DOI: 10.1124/jpet.115.227389] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/25/2015] [Indexed: 01/01/2023] Open
Abstract
Acetaminophen (APAP) is the most commonly used over-the-counter analgesic. However, hepatotoxicity induced by APAP is a major clinical issue, and the factors that define sensitivity to APAP remain unclear. We have previously demonstrated that mice nulled for glutathione S-transferase Pi (GSTP) are resistant to APAP-induced hepatotoxicity. This study aims to exploit this difference to delineate pathways of importance in APAP toxicity. We used mice nulled for GSTP and heme oxygenase-1 oxidative stress reporter mice, together with a novel nanoflow liquid chromatography-tandem mass spectrometry methodology to investigate the role of oxidative stress, cell signaling, and protein S-glutathionylation in APAP hepatotoxicity. We provide evidence that the sensitivity difference between wild-type and Gstp1/2(-/-) mice is unrelated to the ability of APAP to induce oxidative stress, despite observing significant increases in c-Jun N-terminal kinase and extracellular signal-regulated kinase phosphorylation in wild-type mice. The major difference in response to APAP was in the levels of protein S-glutathionylation: Gstp1/2(-/-) mice exhibited a significant increase in the number of S-glutathionylated proteins compared with wild-type animals. Remarkably, these S-glutathionylated proteins are involved in oxidative phosphorylation, respiratory complexes, drug metabolism, and mitochondrial apoptosis. Furthermore, we found that S-glutathionylation of the rate-limiting glutathione-synthesizing enzyme, glutamate cysteine ligase, was markedly increased in Gstp1/2(-/-) mice in response to APAP. The data demonstrate that S-glutathionylation provides an adaptive response to APAP and, as a consequence, suggest that this is an important determinant in APAP hepatotoxicity. This work identifies potential novel avenues associated with cell survival for the treatment of chemical-induced hepatotoxicity.
Collapse
Affiliation(s)
- David J McGarry
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - Probir Chakravarty
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - C Roland Wolf
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - Colin J Henderson
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| |
Collapse
|
91
|
Caterino M, Pastore A, Strozziero MG, Di Giovamberardino G, Imperlini E, Scolamiero E, Ingenito L, Boenzi S, Ceravolo F, Martinelli D, Dionisi-Vici C, Ruoppolo M. The proteome of cblC defect: in vivo elucidation of altered cellular pathways in humans. J Inherit Metab Dis 2015; 38:969-79. [PMID: 25585586 DOI: 10.1007/s10545-014-9806-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 12/26/2022]
Abstract
Methylmalonic acidemia with homocystinuria, cobalamin deficiency type C (cblC) (MMACHC) is the most common inborn error of cobalamin metabolism. Despite a multidrug treatment, the long-term follow-up of early-onset patients is often unsatisfactory, with progression of neurological and ocular impairment. Here, the in-vivo proteome of control and MMACHC lymphocytes (obtained from patients under standard treatment with OHCbl, betaine, folate and L-carnitine) was quantitatively examined by two dimensional differential in-gel electrophoresis (2D-DIGE) and mass spectrometry. Twenty three proteins were found up-regulated and 38 proteins were down-regulated. Consistent with in vivo studies showing disturbance of glutathione metabolism, a deregulation in proteins involved in cellular detoxification, especially in glutathione metabolism was found. In addition, relevant changes were observed in the expression levels of proteins involved in intracellular trafficking and protein folding, energy metabolism, cytoskeleton organization and assembly. This study demonstrates relevant changes in the proteome profile of circulating lymphocytes isolated from treated cblC patients. Some results confirm previous observations in vivo on fibroblast, thus concluding that some dysregulation is ubiquitous. On the other hand, new findings could be tissue-specific. These observations expand our current understanding of the cblC disease and may ignite new research and therapeutic strategies to treat this disorder.
Collapse
|
92
|
Scian M, Atkins WM. The busulfan metabolite EdAG irreversibly glutathionylates glutaredoxins. Arch Biochem Biophys 2015; 583:96-104. [PMID: 26278353 DOI: 10.1016/j.abb.2015.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 01/05/2023]
Abstract
The DNA alkylating agent busulfan is used to 'precondition' patients with leukemia, lymphomas and other hematological disorders prior to hematopoietic stem cell transplants. Busulfan is metabolized via conjugation with glutathione (GSH) followed by intramolecular rearrangement to the GSH analog γ-glutamyl-dehydroalanyl -glycine (EdAG). EdAG contains the electrophilic dehydroalanine, which is expected to react with protein nucleophiles, particularly proteins with GSH binding sites such as glutaredoxins (Grx's). Incubation of EdAG with human Grx-1 or Grx-2 results in facile adduction of cys-23 and cys-77, respectively, as determined by ESI-MS/MS. The resulting modified proteins are catalytically inactive. In contrast, the glutathione transferase A1-1 includes a GSH binding site with a potentially reactive tyrosinate (Tyr-9) but it does not react with EdAG. Similarly, Cys-112 of GSTA1-1, which lies outside the active site and is known to form disulfides with GSH, does not react with EdAG. The results provide the first demonstration of the reactivity of any busulfan metabolites with intact proteins, and they suggest that GSH-binding sites containing thiolates are most susceptible. The adduction of Grx's by EdAG suggests the possible alteration of proteins that are normally regulated via Grx-dependent reversible glutathionylation or deglutathionylation. Dysregulation of Grx-dependent processes could contribute to cellular toxicity of busulfan.
Collapse
Affiliation(s)
- Michele Scian
- The Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, WA 98195-7610, USA
| | - William M Atkins
- The Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, WA 98195-7610, USA.
| |
Collapse
|
93
|
Menon D, Coll R, O'Neill LAJ, Board PG. GSTO1-1 modulates metabolism in macrophages activated through the LPS and TLR4 pathway. J Cell Sci 2015; 128:1982-90. [DOI: 10.1242/jcs.167858] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/10/2015] [Indexed: 02/01/2023] Open
Abstract
ABSTRACT
Macrophages mediate innate immune responses that recognise foreign pathogens, and bacterial lipopolysaccharide (LPS) recruits a signalling pathway through Toll-like receptor 4 (TLR4) to induce pro-inflammatory cytokines and reactive oxygen species (ROS). LPS activation also skews the metabolism of macrophages towards a glycolytic phenotype. Here, we demonstrate that the LPS-triggered glycolytic switch is significantly attenuated in macrophages deficient for glutathione transferase omega-1 (GSTO1, note that GSTO1-1 refers to the dimeric molecule with identical type 1 subunits). In response to LPS, GSTO1-1-deficient macrophages do not produce excess lactate, or dephosphorylate AMPK, a key metabolic stress regulator. In addition, GSTO1-1-deficient cells do not induce HIF1α, which plays a key role in maintaining the pro-inflammatory state of activated macrophages. The accumulation of the TCA cycle intermediates succinate and fumarate that occurs in LPS-treated macrophages was also blocked in GSTO1-1-deficient cells. These data indicate that GSTO1-1 is required for LPS-mediated signalling in macrophages and that it acts early in the LPS–TLR4 pro-inflammatory pathway.
Collapse
Affiliation(s)
- Deepthi Menon
- Department of Molecular Biosciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600, Australia
| | - Rebecca Coll
- Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| | - Luke A. J. O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Philip G. Board
- Department of Molecular Biosciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600, Australia
| |
Collapse
|
94
|
GSTO1*C/GSTO2*G haplotype is associated with risk of transitional cell carcinoma of urinary bladder. Int Urol Nephrol 2015; 47:625-30. [PMID: 25716313 DOI: 10.1007/s11255-015-0933-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/14/2015] [Indexed: 02/01/2023]
Abstract
PURPOSE To clarify the role of genetic polymorphisms of GSTO1 (rs4925) and GSTO2 (rs156697) in individual susceptibility to urinary bladder cancer. METHODS Case-control study consisting of 187 patients with histologically confirmed transitional cell carcinoma (TCC) of urinary bladder and 140 age- and gender-matched cancer-free controls was carried out. Genotyping of GSTO1 and GSTO2 was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). RESULTS We found that carriers of mutant GSTO2*G/G genotype were at increased risk of the development of TCC (OR 2.6, 95% CI 1.2-5.8, p = 0.041), while GSTO1 rs4925 polymorphism was not significantly associated with TCC risk (p = 0.450). According to smoking status, smokers with GSTO2*G/G genotype had significantly higher risk of TCC of urinary bladder (OR 4.3, 95% CI 1.6-11.2, p = 0.003) compared to wild-type carriers with no smoking history. We further analyzed the effects of GSTO1/GSTO2 haplotypes on TCC risk, based on the linkage disequilibrium found for GSTO1 (rs4925) and GSTO2 (rs156697) (D' = 0.309, p = 0.001). The study subjects with GSTO1*C/GSTO2*G (GSTO1 wild-type/GSTO2 mutant) haplotype were at the highest risk of the development of transitional cell carcinoma of urinary bladder (OR 2.8, 95% CI 1.5-5.2, p = 0.002). CONCLUSIONS Our results indicate that GSTO1*C/GSTO2*G haplotype is associated with increased risk of TCC. The modifying effect of GSTO2*G/G genotype on individual susceptibility to TCC is more pronounced, when associated with smoking.
Collapse
|
95
|
Al Khamici H, Brown LJ, Hossain KR, Hudson AL, Sinclair-Burton AA, Ng JPM, Daniel EL, Hare JE, Cornell BA, Curmi PMG, Davey MW, Valenzuela SM. Members of the chloride intracellular ion channel protein family demonstrate glutaredoxin-like enzymatic activity. PLoS One 2015; 10:e115699. [PMID: 25581026 PMCID: PMC4291220 DOI: 10.1371/journal.pone.0115699] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/26/2014] [Indexed: 01/07/2023] Open
Abstract
The Chloride Intracellular Ion Channel (CLIC) family consists of six evolutionarily conserved proteins in humans. Members of this family are unusual, existing as both monomeric soluble proteins and as integral membrane proteins where they function as chloride selective ion channels, however no function has previously been assigned to their soluble form. Structural studies have shown that in the soluble form, CLIC proteins adopt a glutathione S-transferase (GST) fold, however, they have an active site with a conserved glutaredoxin monothiol motif, similar to the omega class GSTs. We demonstrate that CLIC proteins have glutaredoxin-like glutathione-dependent oxidoreductase enzymatic activity. CLICs 1, 2 and 4 demonstrate typical glutaredoxin-like activity using 2-hydroxyethyl disulfide as a substrate. Mutagenesis experiments identify cysteine 24 as the catalytic cysteine residue in CLIC1, which is consistent with its structure. CLIC1 was shown to reduce sodium selenite and dehydroascorbate in a glutathione-dependent manner. Previous electrophysiological studies have shown that the drugs IAA-94 and A9C specifically block CLIC channel activity. These same compounds inhibit CLIC1 oxidoreductase activity. This work for the first time assigns a functional activity to the soluble form of the CLIC proteins. Our results demonstrate that the soluble form of the CLIC proteins has an enzymatic activity that is distinct from the channel activity of their integral membrane form. This CLIC enzymatic activity may be important for protecting the intracellular environment against oxidation. It is also likely that this enzymatic activity regulates the CLIC ion channel function.
Collapse
Affiliation(s)
- Heba Al Khamici
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Louise J. Brown
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Khondker R. Hossain
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Bragg Institute, Australian Nuclear Science and Technology Organisation, Sydney, New South Wales 2234, Australia
| | - Amanda L. Hudson
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Alxcia A. Sinclair-Burton
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Jane Phui Mun Ng
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Elizabeth L. Daniel
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Joanna E. Hare
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bruce A. Cornell
- Surgical Diagnostics, Roseville, Sydney, New South Wales 2069, Australia
| | - Paul M. G. Curmi
- School of Physics, University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Mary W. Davey
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stella M. Valenzuela
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Health Technologies, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
96
|
A platelet protein biochip rapidly detects an Alzheimer's disease-specific phenotype. Acta Neuropathol 2014; 128:665-77. [PMID: 25248508 PMCID: PMC4201753 DOI: 10.1007/s00401-014-1341-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease (AD), a multifactorial neurodegenerative condition caused by genetic and environmental factors, is diagnosed using neuropsychological tests and brain imaging; molecular diagnostics are not routinely applied. Studies have identified AD-specific cerebrospinal fluid (CSF) biomarkers but sample collection requires invasive lumbar puncture. To identify AD-modulated proteins in easily accessible blood platelets, which share biochemical signatures with neurons, we compared platelet lysates from 62 AD, 24 amnestic mild cognitive impairment (aMCI), 13 vascular dementia (VaD), and 12 Parkinson's disease (PD) patients with those of 112 matched controls by fluorescence two-dimensional differential gel electrophoresis in independent discovery and verification sets. The optimal sum score of four mass spectrometry (MS)-identified proteins yielded a sensitivity of 94 % and a specificity of 89 % (AUC = 0.969, 95 % CI = 0.944-0.994) to differentiate AD patients from healthy controls. To bridge the gap between bench and bedside, we developed a high-throughput multiplex protein biochip with great potential for routine AD screening. For convenience and speed of application, this array combines loading control-assisted protein quantification of monoamine oxidase B and tropomyosin 1 with protein-based genotyping for single nucleotide polymorphisms (SNPs) in the apolipoprotein E and glutathione S-transferase omega 1 genes. Based on minimally invasive blood drawing, this innovative protein biochip enables identification of AD patients with an accuracy of 92 % in a single analytical step in less than 4 h.
Collapse
|
97
|
Németi B, Poór M, Gregus Z. A high-performance liquid chromatography-based assay of glutathione transferase omega 1 supported by glutathione or non-physiological reductants. Anal Biochem 2014; 469:12-8. [PMID: 25283130 DOI: 10.1016/j.ab.2014.09.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/11/2014] [Accepted: 09/24/2014] [Indexed: 11/25/2022]
Abstract
The unusual glutathione S-transferase GSTO1 reduces, rather than conjugates, endo- and xenobiotics, and its role in diverse cellular processes has been proposed. GSTO1 has been assayed spectrophotometrically by measuring the disappearance of its substrate, S-(4-nitrophenacyl)glutathione (4-NPG), in the presence of 2-mercaptoethanol that regenerates GSTO1 from its mixed disulfide. To assay GSTO1 in rat liver cytosol, we have developed a high-performance liquid chromatography (HPLC)-based procedure with two main advantages: (i) it measures the formation of the 4-NPG reduction product 4-nitroacetophenone, thereby offering improved sensitivity and accuracy, and (ii) it can use glutathione, the physiological reductant of GSTO1, which is impossible to do with the spectrophotometric procedure. Using the new assay, we show that (i) the GSTO1-catalyzed reduction of 4-NPG in rat liver cytosol also yields 1-(4-nitrophenyl)ethanol, whose formation from 4-nitroacetophenone requires NAD(P)H; (ii) the two assays measure comparable activities with 2-mercaptoethanol or tris(2-carboxyethyl)phosphine used as reductant; (iii) the cytosolic reduction of 4-NPG is inhibited by GSTO1 inhibitors (KT53, 5-chloromethylfluorescein diacetate, and zinc), although the inhibitory effect is strikingly influenced by the type of reductant in the assay and by the sequence of reductant and inhibitor addition. Characterization of GSTO1 inhibitors with the improved assay provides better understanding of interaction of these chemicals with the enzyme.
Collapse
Affiliation(s)
- Balázs Németi
- Department of Pharmacology and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, H-7624 Pécs, Hungary
| | - Miklós Poór
- Department of Pharmacology and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, H-7624 Pécs, Hungary
| | - Zoltán Gregus
- Department of Pharmacology and Pharmacotherapy, Toxicology Section, University of Pécs, Medical School, H-7624 Pécs, Hungary.
| |
Collapse
|
98
|
Abstract
The interaction between antioxidant glutathione and the free thiol in susceptible cysteine residues of proteins leads to reversible protein S-glutathionylation. This reaction ensures cellular homeostasis control (as a common redox-dependent post-translational modification associated with signal transduction) and intervenes in oxidative stress-related cardiovascular pathology (as initiated by redox imbalance). The purpose of this review is to evaluate the recent knowledge on protein S-glutathionylation in terms of chemistry, broad cellular intervention, specific quantification, and potential for therapeutic exploitation. The data bases searched were Medline and PubMed, from 2009 to 2014 (term: glutathionylation). Protein S-glutathionylation ensures protection of protein thiols against irreversible over-oxidation, operates as a biological redox switch in both cell survival (influencing kinases and protein phosphatases pathways) and cell death (by potentiation of apoptosis), and cross-talks with phosphorylation and with S-nitrosylation. Collectively, protein S-glutathionylation appears as a valuable biomarker for oxidative stress, with potential for translation into novel therapeutic strategies.
Collapse
Affiliation(s)
- Doina Popov
- Institute of Cellular Biology and Pathology "N. Simionescu" of the Romanian Academy , 8, B.P. Hasdeu Street, Bucharest 050568 , Romania
| |
Collapse
|
99
|
Lallement PA, Brouwer B, Keech O, Hecker A, Rouhier N. The still mysterious roles of cysteine-containing glutathione transferases in plants. Front Pharmacol 2014; 5:192. [PMID: 25191271 PMCID: PMC4138524 DOI: 10.3389/fphar.2014.00192] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/26/2014] [Indexed: 12/31/2022] Open
Abstract
Glutathione transferases (GSTs) represent a widespread multigenic enzyme family able to modify a broad range of molecules. These notably include secondary metabolites and exogenous substrates often referred to as xenobiotics, usually for their detoxification, subsequent transport or export. To achieve this, these enzymes can bind non-substrate ligands (ligandin function) and/or catalyze the conjugation of glutathione onto the targeted molecules, the latter activity being exhibited by GSTs having a serine or a tyrosine as catalytic residues. Besides, other GST members possess a catalytic cysteine residue, a substitution that radically changes enzyme properties. Instead of promoting GSH-conjugation reactions, cysteine-containing GSTs (Cys-GSTs) are able to perform deglutathionylation reactions similarly to glutaredoxins but the targets are usually different since glutaredoxin substrates are mostly oxidized proteins and Cys-GST substrates are metabolites. The Cys-GSTs are found in most organisms and form several classes. While Beta and Omega GSTs and chloride intracellular channel proteins (CLICs) are not found in plants, these organisms possess microsomal ProstaGlandin E-Synthase type 2, glutathionyl hydroquinone reductases, Lambda, Iota and Hemerythrin GSTs and dehydroascorbate reductases (DHARs); the four last classes being restricted to the green lineage. In plants, whereas the role of DHARs is clearly associated to the reduction of dehydroascorbate to ascorbate, the physiological roles of other Cys-GSTs remain largely unknown. In this context, a genomic and phylogenetic analysis of Cys-GSTs in photosynthetic organisms provides an updated classification that is discussed in the light of the recent literature about the functional and structural properties of Cys-GSTs. Considering the antioxidant potencies of phenolic compounds and more generally of secondary metabolites, the connection of GSTs with secondary metabolism may be interesting from a pharmacological perspective.
Collapse
Affiliation(s)
- Pierre-Alexandre Lallement
- UMR1136, Interactions Arbres - Microorganismes, Université de Lorraine Vandoeuvre-lès-Nancy, France ; UMR1136, Interactions Arbres - Microorganismes, INRA Champenoux, France
| | - Bastiaan Brouwer
- Department of Plant Physiology, Umeå Plant Science Centre, Umeå University Umeå, Sweden
| | - Olivier Keech
- Department of Plant Physiology, Umeå Plant Science Centre, Umeå University Umeå, Sweden
| | - Arnaud Hecker
- UMR1136, Interactions Arbres - Microorganismes, Université de Lorraine Vandoeuvre-lès-Nancy, France ; UMR1136, Interactions Arbres - Microorganismes, INRA Champenoux, France
| | - Nicolas Rouhier
- UMR1136, Interactions Arbres - Microorganismes, Université de Lorraine Vandoeuvre-lès-Nancy, France ; UMR1136, Interactions Arbres - Microorganismes, INRA Champenoux, France
| |
Collapse
|
100
|
Menon D, Coll R, O'Neill LAJ, Board PG. Glutathione transferase omega 1 is required for the lipopolysaccharide-stimulated induction of NADPH oxidase 1 and the production of reactive oxygen species in macrophages. Free Radic Biol Med 2014; 73:318-27. [PMID: 24873723 DOI: 10.1016/j.freeradbiomed.2014.05.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 11/19/2022]
Abstract
Bacterial lipopolysaccharide (LPS) stimulation of macrophages and inflammation via the Toll-like receptor 4 (TLR4) signaling pathway through NF-κΒ generates reactive oxygen species (ROS) and proinflammatory cytokines such as IL-1β, IL-6, and TNFα. Because glutathione transferase Omega 1-1 (GSTO1-1) can catalyze redox reactions such as the deglutathionylation of proteins and has also been implicated in the release of IL-1β we investigated its role in the development of LPS-mediated inflammation. Our data show that shRNA knockdown of GSTO1-1 in macrophage-like J774.1A cells blocks the expression of NADPH oxidase 1 and the generation of ROS after LPS stimulation. Similar results were obtained with a GSTO1-1 inhibitor. To maintain high ROS levels during an inflammatory response, LPS stimulation causes the suppression of enzymes such as catalase and glutathione peroxidase that protect against oxidative stress. The knockdown of GSTO1-1 also attenuates this response. Our data indicate that GSTO1-1 needs to be catalytically active and mediates its effects on the LPS/TLR4 inflammatory pathway upstream of NF-κΒ. These data suggest that GSTO1-1 is a novel target for anti-inflammatory intervention.
Collapse
Affiliation(s)
- Deepthi Menon
- Department of Molecular Biosciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600, Australia
| | - Rebecca Coll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin 2, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin 2, Ireland
| | - Philip G Board
- Department of Molecular Biosciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|