1
|
Bruschi M, Biancucci F, Masini S, Piacente F, Ligi D, Bartoccini F, Antonelli A, Mannello F, Bruzzone S, Menotta M, Fraternale A, Magnani M. The influence of redox modulation on hypoxic endothelial cell metabolic and proteomic profiles through a small thiol-based compound tuning glutathione and thioredoxin systems. Biofactors 2023; 49:1205-1222. [PMID: 37409789 DOI: 10.1002/biof.1988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/16/2023] [Indexed: 07/07/2023]
Abstract
Reduction in oxygen levels is a key feature in the physiology of the bone marrow (BM) niche where hematopoiesis occurs. The BM niche is a highly vascularized tissue and endothelial cells (ECs) support and regulate blood cell formation from hematopoietic stem cells (HSCs). While in vivo studies are limited, ECs when cultured in vitro at low O2 (<5%), fail to support functional HSC maintenance due to oxidative environment. Therefore, changes in EC redox status induced by antioxidant molecules may lead to alterations in the cellular response to hypoxia likely favoring HSC self-renewal. To evaluate the impact of redox regulation, HUVEC, exposed for 1, 6, and 24 h to 3% O2 were treated with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152). Metabolomic analyses revealed that I-152 increased glutathione levels and influenced the metabolic profiles interconnected with the glutathione system and the redox couples NAD(P)+/NAD(P)H. mRNA analysis showed a lowered gene expression of HIF-1α and VEGF following I-152 treatment whereas TRX1 and 2 were stimulated. Accordingly, the proteomic study revealed the redox-dependent upregulation of thioredoxin and peroxiredoxins that, together with the glutathione system, are the main regulators of intracellular ROS. Indeed, a time-dependent ROS production under hypoxia and a quenching effect of the molecule were evidenced. At the secretome level, the molecule downregulated IL-6, MCP-1, and PDGF-bb. These results suggest that redox modulation by I-152 reduces oxidative stress and ROS level in hypoxic ECs and may be a strategy to fine-tune the environment of an in vitro BM niche able to support functional HSC maintenance.
Collapse
Affiliation(s)
- Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Federica Biancucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Sofia Masini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genoa, Genoa, GE, Italy
| | - Daniela Ligi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Francesca Bartoccini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Ferdinando Mannello
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genoa, Genoa, GE, Italy
- IRCCS, Ospedale Policlinico San Martino, Genoa, GE, Italy
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| |
Collapse
|
2
|
Zhang J, Ye ZW, Morgenstern R, Townsend DM, Tew KD. Microsomal glutathione transferase 1 in cancer and the regulation of ferroptosis. Adv Cancer Res 2023; 160:107-132. [PMID: 37704286 PMCID: PMC10586476 DOI: 10.1016/bs.acr.2023.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Microsomal glutathione transferase 1 (MGST1) is a member of the MAPEG family (membrane associated proteins in eicosanoid and glutathione metabolism), defined according to enzymatic activities, sequence motifs, and structural properties. MGST1 is a homotrimer which can bind three molecules of glutathione (GSH), with one modified to a thiolate anion displaying one-third-of-sites-reactivity. MGST1 has both glutathione transferase and peroxidase activities. Each is based on stabilizing the GSH thiolate in the same active site. MGST1 is abundant in the liver and displays a broad subcellular distribution with high levels in endoplasmic reticulum and mitochondrial membranes, consistent with a physiological role in protection from reactive electrophilic intermediates and oxidative stress. In this review paper, we particularly focus on recent advances made in understanding MGST1 activation, induction, broad subcellular distribution, and the role of MGST1 in apoptosis, ferroptosis, cancer progression, and therapeutic responses.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Ralf Morgenstern
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Danyelle M Townsend
- Department of Drug Discovery 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
|
3
|
Xue XL, Zhang H, Chen GH, Yu GH, Hu HR, Niu SY, Wang KP, Hu ZQ. Coumarin-cyanine hybrid: A ratiometric fluorescent probe for accurate detection of peroxynitrite in mitochondria. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 292:122443. [PMID: 36753868 DOI: 10.1016/j.saa.2023.122443] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/22/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
There is an urgent need to develop highly sensitive and selective fluorescence probes for ONOO- in mitochondria. Herein, we reported a ratiometric fluorescent probe COUS with coumarin-cyanine hybrid as fluorophore and C = C bonds as reaction sites of ONOO-. The probe COUS was sensitive and selective to ONOO-, and had a large fluorescence emission shift (239 nm) as well as a low detection limit (41.88 nM). Moreover, COUS showed the mitochondrial targeting ability, and the targeting moiety could dissociate from the probe when reacting with ONOO-, which enabled COUS to accurately detect ONOO- in mitochondria.
Collapse
Affiliation(s)
- Xiao-Lei Xue
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Hao Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Gui-Hua Chen
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Guan-Hua Yu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Hao-Ran Hu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shu-Yan Niu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Kun-Peng Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Zhi-Qiang Hu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
4
|
Samarin J, Fabrowski P, Kurilov R, Nuskova H, Hummel-Eisenbeiss J, Pink H, Li N, Weru V, Alborzinia H, Yildiz U, Grob L, Taubert M, Czech M, Morgen M, Brandstädter C, Becker K, Mao L, Jayavelu AK, Goncalves A, Uhrig U, Seiler J, Lyu Y, Diederichs S, Klingmüller U, Muckenthaler M, Kopp-Schneider A, Teleman A, Miller AK, Gunkel N. Low level of antioxidant capacity biomarkers but not target overexpression predicts vulnerability to ROS-inducing drugs. Redox Biol 2023; 62:102639. [PMID: 36958250 PMCID: PMC10053401 DOI: 10.1016/j.redox.2023.102639] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Despite a strong rationale for why cancer cells are susceptible to redox-targeting drugs, such drugs often face tumor resistance or dose-limiting toxicity in preclinical and clinical studies. An important reason is the lack of specific biomarkers to better select susceptible cancer entities and stratify patients. Using a large panel of lung cancer cell lines, we identified a set of "antioxidant-capacity" biomarkers (ACB), which were tightly repressed, partly by STAT3 and STAT5A/B in sensitive cells, rendering them susceptible to multiple redox-targeting and ferroptosis-inducing drugs. Contrary to expectation, constitutively low ACB expression was not associated with an increased steady state level of reactive oxygen species (ROS) but a high level of nitric oxide, which is required to sustain high replication rates. Using ACBs, we identified cancer entities with a high percentage of patients with favorable ACB expression pattern, making it likely that more responders to ROS-inducing drugs could be stratified for clinical trials.
Collapse
Affiliation(s)
- Jana Samarin
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Piotr Fabrowski
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roman Kurilov
- Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hana Nuskova
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Hannelore Pink
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nan Li
- Somatic Evolution and Early Detection, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vivienn Weru
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Umut Yildiz
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Grob
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Minerva Taubert
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marie Czech
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Morgen
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Brandstädter
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Lianghao Mao
- Proteomics and Cancer Cell Signaling Group, CCU Pediatric Leukemia, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ashok Kumar Jayavelu
- Proteomics and Cancer Cell Signaling Group, CCU Pediatric Leukemia, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angela Goncalves
- Somatic Evolution and Early Detection, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Uhrig
- Chemical Biology Core Facility, EMBL, Heidelberg, Germany
| | - Jeanette Seiler
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yanhong Lyu
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
| | - Sven Diederichs
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Martina Muckenthaler
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
| | | | - Aurelio Teleman
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aubry K Miller
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Nikolas Gunkel
- Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
5
|
Varešlija D, Tipton KF, Davey GP, McDonald AG. 6-Hydroxydopamine: a far from simple neurotoxin. J Neural Transm (Vienna) 2020; 127:213-230. [DOI: 10.1007/s00702-019-02133-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/21/2019] [Indexed: 12/13/2022]
|
6
|
Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1949-1967. [DOI: 10.1016/j.bbadis.2018.11.007] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 02/06/2023]
|
7
|
Axton ER, Cristobal E, Choi J, Miranda CL, Stevens JF. Metabolomics-Driven Elucidation of Cellular Nitrate Tolerance Reveals Ascorbic Acid Prevents Nitroglycerin-Induced Inactivation of Xanthine Oxidase. Front Pharmacol 2018; 9:1085. [PMID: 30319419 PMCID: PMC6167911 DOI: 10.3389/fphar.2018.01085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/06/2018] [Indexed: 12/16/2022] Open
Abstract
Glyceryl trinitrate (GTN) has found widespread use for the treatment of angina pectoris, a pathological condition manifested by chest pain resulting from insufficient blood supply to the heart. Metabolic conversion of GTN, a nitric oxide (NO) pro-drug, into NO induces vasodilation and improves blood flow. Patients develop tolerance to GTN after several weeks of continuous use, limiting the potential for long-term therapy. The mechanistic cause of nitrate tolerance is relatively unknown. We developed a cell culture model of nitrate tolerance that utilizes stable isotopes to measure metabolism of 15N3-GTN into 15N-nitrite. We performed global metabolomics to identify the mechanism of GTN-induced nitrate tolerance and to elucidate the protective role of vitamin C (ascorbic acid). Metabolomics analyses revealed that GTN impaired purine metabolism and depleted intracellular ATP and GTP. GTN inactivated xanthine oxidase (XO), an enzyme that is critical for the metabolic bioactivation of GTN into NO. Ascorbic acid prevented inactivation of XO, resulting in increased NO production from GTN. Our studies suggest that ascorbic acid has the ability to prevent nitrate tolerance by protecting XO, but not aldehyde dehydrogenase (another GTN bioactivating enzyme), from GTN-induced inactivation. Our findings provide a mechanistic explanation for the previously observed beneficial effects of ascorbic acid in nitrate therapy.
Collapse
Affiliation(s)
- Elizabeth Rose Axton
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States.,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Eleonso Cristobal
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Cristobal L Miranda
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Jan Frederik Stevens
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
8
|
Ferrer-Sueta G, Campolo N, Trujillo M, Bartesaghi S, Carballal S, Romero N, Alvarez B, Radi R. Biochemistry of Peroxynitrite and Protein Tyrosine Nitration. Chem Rev 2018; 118:1338-1408. [DOI: 10.1021/acs.chemrev.7b00568] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gerardo Ferrer-Sueta
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Nicolás Campolo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Silvina Bartesaghi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Carballal
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Romero
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Beatriz Alvarez
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
9
|
Paranjpe A, Srivenugopal KS. Degradation of NF-κB, p53 and other regulatory redox-sensitive proteins by thiol-conjugating and -nitrosylating drugs in human tumor cells. Carcinogenesis 2013; 34:990-1000. [PMID: 23354308 DOI: 10.1093/carcin/bgt032] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The ionized cysteines present on the surfaces of many redox-sensitive proteins play functionally essential roles and are readily targeted by the reactive oxygen and reactive nitrogen species. Using disulfiram (DSF) and nitroaspirin (NCX4016) as the model compounds that mediate thiol-conjugating and nitrosylating reactions, respectively, we investigated the fate of p53, nuclear factor-kappaB (NF-κB) and other redox-responsive proteins following the exposure of human cancer cell lines to the drugs. Both drugs induced glutathionylation of bulk proteins in tumor cells and cell-free extracts. A prominent finding of this study was a time- and dose-dependent degradation of the redox-regulated proteins after brief treatments of tumor cells with DSF or NCX4016. DSF and copper-chelated DSF at concentrations of 50-200 µM induced the disappearance of wild-type p53, mutant p53, NF-κB subunit p50 and the ubiquitin-activating enzyme E1 (UBE1) in tumor cell lines. DSF also induced the glutathionylation of p53. The recombinant p53 protein modified by DSF was preferentially degraded by rabbit reticulocyte lysates. The proteasome inhibitor PS341 curtailed the DSF-induced degradation of p53 in HCT116 cells. Further, the NCX4016 induced a dose-dependent disappearance of the UBE1 and NF-κB p50 proteins in cell lines, besides a time-dependent degradation of aldehyde dehydrogenase in mouse liver after a single injection of 150 mg/kg. The loss of p53 and NF-kB proteins correlated with decreases in their specific binding to DNA. Our results demonstrate the hitherto unrecognized ability of the non-toxic thiolating and nitrosylating agents to degrade regulatory proteins and highlight the exploitable therapeutic benefits.
Collapse
Affiliation(s)
- Ameya Paranjpe
- Department of Biomedical Sciences, Cancer Biology Research Centre, School of Pharmacy, Texas Tech University Health Sciences Center, 1406 S. Coulter Drive, Amarillo, TX 79106, USA
| | | |
Collapse
|
10
|
Tanvir S, Merlier F, Pulvin S. Biosensing of reactive intermediates produced by the photocatalytic activities of titanium dioxide nanoparticles. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2012; 110:22-7. [PMID: 22417814 DOI: 10.1016/j.jphotobiol.2012.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 01/16/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
Abstract
The development of an enzyme based biosensing method is described for evaluating the toxicity of solutions treated by titanium dioxide photocatalysis. The method is based on the potential of rat liver microsomal glutathione transferase ability (mGST) to get enhanced in the conditions of chemical and oxidative toxicity. Phenol is taken as a model pollutant due to its toxicity and prevalence in industrial processes. Chemical analysis of the parent compound, products and acute toxicity assays using the mGST activity, were conducted during and after the various photocatalytic treatments. The maximum mGST activity was observed from 60 and 120 min treated samples. This post-treatment toxicity might be due to toxic phenolic products, which may include p-benzoquinone, hydroquinone, benzenetriol and other intermediates. The enzymatic activity pattern observed after photocatalytic treatment corresponded well with the chemical degradation data obtained by HPLC-UV. The mGST assay seems to be an easy to use and promising approach for evaluating the effectiveness of wastewater treatment processes.
Collapse
Affiliation(s)
- Shazia Tanvir
- Laboratoire de Génie Enzymatique et Cellulaire, UMR 6022 CNRS, Université de Technologie de Compiègne, BP 20529-60205 Compiègne Cedex, France.
| | | | | |
Collapse
|
11
|
Tanvir S, Adenier H, Pulvin S. Screening and prediction of reactive intermediates in a microreactor with immobilized rat hepatic microsomes using acetaminophen as a model drug. Enzyme Microb Technol 2009. [DOI: 10.1016/j.enzmictec.2009.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
12
|
Zaragozá R, Torres L, García C, Eroles P, Corrales F, Bosch A, Lluch A, García-Trevijano ER, Viña JR. Nitration of cathepsin D enhances its proteolytic activity during mammary gland remodelling after lactation. Biochem J 2009; 419:279-288. [PMID: 19125694 DOI: 10.1042/bj20081746] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Proteomic studies in the mammary gland of control lactating and weaned rats have shown that there is an increased pattern of nitrated proteins during weaning when compared with controls. Here we report the novel finding that cathepsin D is nitrated during weaning. The expression and protein levels of this enzyme are increased after 8 h of litter removal and this up-regulation declines 5 days after weaning. However, there is a marked delay in cathepsin D activity since it does not increase until 2 days post-weaning and remains high thereafter. In order to find out whether nitration of cathepsin D regulates its activity, iNOS (inducible nitric oxide synthase)(-/-) mice were used. The expression and protein levels of this enzyme were similar to WT (wild-type) animals, but the proteolytic activity was significantly reduced during weaning in knockout compared to WT mice. in vitro treatment of recombinant human cathepsin D or lactating mammary gland homogenates with relatively low concentrations of peroxynitrite enhances the nitration as well as specific activity of this enzyme. Using MS, it has been shown that the residue Tyr168 was nitrated. All of these results show that protein nitration during weaning might be a signalling pathway involved in mammary gland remodelling.
Collapse
Affiliation(s)
- Rosa Zaragozá
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, E-46010 Valencia, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Contribution of liver mitochondrial membrane-bound glutathione transferase to mitochondrial permeability transition pores. Toxicol Appl Pharmacol 2009; 235:77-85. [DOI: 10.1016/j.taap.2008.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 11/12/2008] [Accepted: 11/17/2008] [Indexed: 12/20/2022]
|
14
|
Abstract
Felinine is a branched-chain sulfur amino acid present in the urine of certain Felidae, including domestic cats. The objective of the present study was to determine if additional cystine and/or dietary N would increase felinine and N-acetylfelinine excretion by intact male cats fed a low-protein (LP) diet. Feeding five adult intact male cats an LP diet (18·8 % of metabolisable energy (ME) as protein) v. a high-protein diet (38·6 % of ME as protein) resulted in a trend (P = 0·08) for decreased urinary felinine and no change in N-acetylfelinine excretion. In a 23 d study, when the LP diet was supplemented with l-cystine at 9·3 g/kg DM, urinary felinine:creatinine ratio showed a linear two-fold (121 %) increase (P < 0·01) from 0·24 (sem 0·05) to 0·53 (sem 0·13) after 10 d. Subsequent feeding of the LP diet resulted in a decrease in felinine excretion to base levels. Plasma γ-glutamylfelinylglycine concentrations were consistent with the excretion of felinine. Supplementation of the LP diet with l-cystine (9·3 g/kg DM), dispensable amino acids and arginine to a second group (n 5) also resulted in a significant (P < 0·01) but smaller (+72 %) increase in the daily felinine:creatinine ratio (0·25 (sem 0·04) to 0·43 (sem 0·05)). The degree of felinine N-acetylation within groups was unaffected by dietary addition and withdrawal of amino acids. The results indicate that felinine synthesis is regulated by cystine availability, and that arginine may be physiologically important in decreasing felinine biosynthesis in intact male cats.
Collapse
|
15
|
Lee KK, Shimoji M, Hossain QS, Sunakawa H, Aniya Y. Novel function of glutathione transferase in rat liver mitochondrial membrane: Role for cytochrome c release from mitochondria☆⁎Some of the data were presented at the ISSX (International Society for Study of Xenobiotics) Meeting, Oct.12, 2007, in Sendai, Japan. Toxicol Appl Pharmacol 2008; 232:109-18. [DOI: 10.1016/j.taap.2008.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 04/30/2008] [Accepted: 06/17/2008] [Indexed: 10/21/2022]
|
16
|
Zhu DY, Du Y, Huang X, Guo MY, Ma KF, Yu YP, Lou YJ. MAPEG Expression in Mouse Embryonic Stem Cell-Derived Hepatic Tissue System. Stem Cells Dev 2008; 17:775-83. [DOI: 10.1089/scd.2007.0241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Dan-Yan Zhu
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Yue Du
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Xin Huang
- Cancer Institute, College of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Zhejiang-California International NanoSystems Institute, Zhejiang University, Hangzhou, China
| | - Mei-Yuan Guo
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Kui-Fen Ma
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, Zhejiang University, Hangzhou, China
| | - Yong-Ping Yu
- Institute of Material Medica, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Jia Lou
- Institute of Pharmacology-Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
McKenzie MJ, Goldfarb AH. Aerobic exercise bout effects on gene transcription in the rat soleus. Med Sci Sports Exerc 2007; 39:1515-21. [PMID: 17805083 DOI: 10.1249/mss.0b013e318074c256] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE This study was designed to identify changes in gene transcription that occur in the soleus muscle of untrained, 10-wk-old rats after a single aerobic exercise bout, and to identify which families of genes are most likely affected. METHODS Rats were either run for 2 h and killed 1 h after exercise, or they remained sedentary and were killed at a matched time. Soleus muscles from each animal were examined using DNA microarrays, four genes related to RONS were analyzed by PCR, and two proteins were checked by Western blot analysis. RESULTS The microarray identified 52 genes significantly altered by the exercise. The major gene families altered were metabolism, apoptosis, muscle contraction, transcription/cell signaling, tissue generation, and inflammation. Real-time PCR was performed on four genes (NFkappaB, TNFalpha, Atf3, and Mgst1), and the results from PCR analysis agreed with the microarray results. NFkappaB and TNFalpha were unaltered, whereas Atf3 was upregulated and Mgst1 was downregulated in the exercised soleus muscles. NFkappaB protein level was not different between the two groups, whereas Atf3 protein level was elevated in the exercise group according to Western blot analysis. CONCLUSIONS These data suggest that 1 h after a 2-h run at approximately 65% of VO2max, the soleus muscle undergoes significant gene-transcript changes. Also, the genes examined with the real-time PCR matched the microarray results and the measured protein concentration concentrations agreed with gene-transcript data at the 1-h postexercise time point.
Collapse
Affiliation(s)
- Michael J McKenzie
- Department of Exercise and Sport Science, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | | |
Collapse
|
18
|
Rebrin I, Brégère C, Kamzalov S, Gallaher TK, Sohal RS. Nitration of tryptophan 372 in succinyl-CoA:3-ketoacid CoA transferase during aging in rat heart mitochondria. Biochemistry 2007; 46:10130-44. [PMID: 17685555 PMCID: PMC2526316 DOI: 10.1021/bi7001482] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The main objective of this study was to test the hypothesis that in vivo post-translational modifications in proteins, induced by the endogenously generated reactive oxygen and nitrogen molecules, can alter protein function and thereby have an effect on metabolic pathways during the aging process. Succinyl-CoA:3-ketoacid coenzyme A transferase (SCOT), the mitochondrial enzyme involved in the breakdown of ketone bodies in the extrahepatic tissues, was identified in rat heart to undergo age-associated increase in a novel, nitro-hydroxy, addition to tryptophan 372, located in close proximity ( approximately 10 A) of the enzyme active site. Between 4 and 24 months of age, the molar content of nitration was more than doubled while specific enzyme activity increased significantly. The amount of SCOT protein, however, remained unchanged. In vitro treatment of heart mitochondrial soluble proteins with relatively low concentrations of peroxynitrite enhanced the nitration as well as specific activity of SCOT. Results of this study identify tryptophan to be a specific target of nitration in vivo, for the first time. We hypothesize that increases in tryptophan nitration of SCOT and catalytic activity constitute a plausible mechanism for the age-related metabolic shift toward enhanced ketone body consumption as an alternative source of energy supply in the heart.
Collapse
Affiliation(s)
- Igor Rebrin
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|
19
|
Siritantikorn A, Johansson K, Ahlen K, Rinaldi R, Suthiphongchai T, Wilairat P, Morgenstern R. Protection of cells from oxidative stress by microsomal glutathione transferase 1. Biochem Biophys Res Commun 2007; 355:592-6. [PMID: 17306223 DOI: 10.1016/j.bbrc.2007.02.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Accepted: 02/03/2007] [Indexed: 12/31/2022]
Abstract
Rat liver microsomal glutathione transferase 1 (MGST1) is a membrane-bound enzyme that displays both glutathione transferase and glutathione peroxidase activities. We hypothesized that physiologically relevant levels of MGST1 is able to protect cells from oxidative damage by lowering intracellular hydroperoxide levels. Such a role of MGST1 was studied in human MCF7 cell line transfected with rat liver mgst1 (sense cell) and with antisense mgst1 (antisense cell). Cytotoxicities of two hydroperoxides (cumene hydroperoxide (CuOOH) and hydrogen peroxide) were determined in both cell types using short-term and long-term cytotoxicity assays. MGST1 significantly protected against CuOOH and against hydrogen peroxide (although less pronounced and only in short-term tests). These results demonstrate that MGST1 can protect cells from both lipophilic and hydrophilic hydroperoxides, of which only the former is a substrate. After CuOOH exposure MGST1 significantly lowered intracellular ROS as determined by FACS analysis.
Collapse
Affiliation(s)
- Atchasai Siritantikorn
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | | | | | | | | | | |
Collapse
|
20
|
Brito PM, Mariano A, Almeida LM, Dinis TCP. Resveratrol affords protection against peroxynitrite-mediated endothelial cell death: A role for intracellular glutathione. Chem Biol Interact 2006; 164:157-66. [PMID: 17078938 DOI: 10.1016/j.cbi.2006.09.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 09/14/2006] [Accepted: 09/19/2006] [Indexed: 10/24/2022]
Abstract
Atherosclerosis, the main cause of cardiovascular disease (CD), is a chronic inflammatory condition associated with an overproduction of oxidant species, namely peroxynitrite, which is a powerful oxidant that reacts directly with all biomolecules. Glutathione is an efficient scavenger of peroxynitrite, so, modulation of glutathione synthesis may provide a strategy to selectively protect cells from this oxidant. Here, we investigated the ability of resveratrol, a component of red wine, to prevent peroxynitrite-mediated endothelial cells toxicity and the underlying mechanism. Bovine aortic endothelial cells (BAEC) in primary cultures were treated with authentic peroxynitrite and the cell viability and intracellular glutathione contents were assessed. Our results demonstrate that a long pre-incubation (14 h) of BAEC with resveratrol (1-50 microM) leads to the endothelial cells rescue from injury triggered by authentic peroxynitrite by a mechanism of up-regulation of the intracellular GSH content, for the highest resveratrol concentration tested. Considering the importance of GSH in regulation of cell life, this capacity of resveratrol provides a new mechanism for its cardioprotective effects and may contribute to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Paula M Brito
- Laboratório de Bioquímica, Faculdade de Farmácia, Universidade de Coimbra and Centro de Neurociências, Universidade de Coimbra, 3000-295 Coimbra, Portugal
| | | | | | | |
Collapse
|
21
|
Ji Y, Bennett BM. Biotransformation of glyceryl trinitrate by rat hepatic microsomal glutathione S-transferase 1. J Pharmacol Exp Ther 2006; 318:1050-6. [PMID: 16720755 DOI: 10.1124/jpet.106.103713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although the biotransformation of organic nitrates by the cytosolic glutathione S-transferases (GSTs) is well known, the relative contribution of the microsomal GST (MGST1) to nitrate biotransformation has not been described. We therefore compared the denitration of glyceryl trinitrate (GTN) by purified rat liver MGST1 and cytosolic GSTs. Both MGST1 and cytosolic GSTs catalyzed the denitration of GTN, but the activity of MGST1 toward GTN was 2- to 3-fold higher. To mimic oxidative/nitrosative stress in vitro, we treated enzyme preparations with hydrogen peroxide, S-nitrosoglutathione, and peroxynitrite. Both oxidants and nitrating reagents increased the activity of MGST1 toward the GST substrate, 1-chloro-2,4-dinitrobenzene (CDNB) whereas these treatments inhibited GTN denitration by MGST1. Alkylation of the sole cysteine residue of MGST1 by N-ethylmaleimide markedly increased enzyme activity with CDNB as substrate but decreased the rate of GTN denitration. In aortic microsomes from GTN-tolerant animals, there was a decreased abundance of MGST1 dimers and trimers. In hepatic microsomes from GTN-tolerant animals, GTN biotransformation was unaltered whereas the rate of CDNB conjugation was doubled, suggesting that chronic GTN exposure causes structural modifications to the enzyme, resulting in increased activity to certain substrates. Collectively, these data indicate that MGST1 contributes significantly to the biotransformation of GTN and that chemical modification of the microsomal enzyme has differential effects on the catalytic activity toward different substrates.
Collapse
Affiliation(s)
- Yanbin Ji
- Department of Pharmacology and Toxicology, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada K7L 3N6
| | | |
Collapse
|
22
|
Shi Q, Chen HF, Lou YJ. Further evidence that rat liver microsomal glutathione transferase 1 is not a cellular protein target for S-nitrosylation. Chem Biol Interact 2006; 162:228-36. [PMID: 16899233 DOI: 10.1016/j.cbi.2006.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 06/26/2006] [Accepted: 06/29/2006] [Indexed: 11/25/2022]
Abstract
By adopting biotin switch method, we recently reported that liver microsomal glutathione transferase 1 (MGST1) might not be a protein target for S-nitrosylation in rat microsomes or in vivo. However, alternative analytic methods are needed to confirm this observation, as a single biotin switch method in judging specific protein S-nitrosylation in biological samples is increasingly recognized as insufficient, or even unreliable. Besides, only MGST1 localized on endoplasmic reticulum (ER), but not mitochondria which favors protein S-nitrosylation was examined in the previous report. Present study was therefore carried out to address these issues. Primary cultured hepatocytes were used. A physiological existing nitric oxide (NO) donor S-nitrosoglutathione (GSNO) was adopted to trigger protein S-nitrosylation. MGST1 was immunoprecipitated and its S-nitrosothiol content was measured by the NO probe 2,3-diaminonaphthalene. In parallel, S-nitrosylated proteins were immunoprecipitated by a monoclonal anti-S-nitrosocysteine antibody and probed with an anti-MGST1 antibody. In hepatocytes, neither ER nor mitochondria were found to contain S-nitrosylated MGST1 after GSNO treatment, showing that differently distributed MGST1 was consistently un-nitrosylable in the cellular environment. But under broken cell conditions, when samples were incubated directly with GSNO, MGST1 S-nitrosylation was indeed detectable in both the microsomal and mitochondrial proteins, indicating that previous failure in detecting MGST1 S-nitrosylation in microsomes is due to the limitations of biotin switch method. These results clearly, if not definitely, demonstrate that MGST1 is not a ready candidate for S-nitrosylation in the cellular content, despite its susceptibility to S-nitrosylation under broken cell conditions.
Collapse
Affiliation(s)
- Qiang Shi
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310031, China
| | | | | |
Collapse
|
23
|
Imaizumi N, Miyagi S, Aniya Y. Reactive nitrogen species derived activation of rat liver microsomal glutathione S-transferase. Life Sci 2006; 78:2998-3006. [PMID: 16386761 DOI: 10.1016/j.lfs.2005.11.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2005] [Revised: 11/16/2005] [Accepted: 11/25/2005] [Indexed: 11/25/2022]
Abstract
The effect of reactive nitrogen species on rat liver microsomal glutathione S-transferase (MGST1) was investigated using microsomes and purified MGST1. When microsomes or the purified enzyme were incubated with peroxynitrite (ONOO(-)), the GST activity was increased to 2.5-6.5 fold in concentration-dependent manner and a small amount of the MGST1 dimer was detected. MGST1 activity was increased by ONOO(-) in the presence of high amounts of reducing agents including glutathione (GSH) and the activities increased by ONOO(-) or ONOO(-) plus GSH treatment were decreased by 30-40% by further incubation with dithiothreitol (DTT, reducing disulfide) or by sodium arsenite (reducing sulfenic acid). Furthermore, GSH was detected by HPLC from the MGST1 which was incubated with ONOO(-) plus GSH or S-nitrosoglutathione followed by DTT treatment. In addition, the MGST1 activity increased by nitric oxide (NO) donors such as S-nitrosoglutathione, S-nitrosocysteine or the non-thiol NO donor 1-hydroxy-2-oxo-3 (3-aminopropyl)-3-isopropyl was restored by the DTT treatment. Since DTT can reduce S-nitrosothiol and disulfide bond to thiol, S-nitrosylation and a mixed disulfide bond formation of MGST1 were suggested. Thus, it was demonstrated that MGST1 is activated by reactive nitrogen species through a forming dimeric protein, mixed disulfide bond, nitrosylation and sulfenic acid.
Collapse
Affiliation(s)
- Naoki Imaizumi
- Laboratory of Functional and Molecular Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| | | | | |
Collapse
|
24
|
Zhao Y, Li H, Gao Z, Gong Y, Xu H. Effects of flavonoids extracted from Scutellaria baicalensis Georgi on hemin–nitrite–H2O2 induced liver injury. Eur J Pharmacol 2006; 536:192-9. [PMID: 16574098 DOI: 10.1016/j.ejphar.2006.02.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Revised: 02/10/2006] [Accepted: 02/21/2006] [Indexed: 01/01/2023]
Abstract
Hemin-nitrite-H2O2 system may play a role in liver oxidative injury in some pathological events. In this paper, the effects of the three active components of the root of Scutellaria baicalensis Georgi, i.e. baicalin, baicalein and wogonin, on hemin-nitrite-H2O2 induced liver injury were studied in liver homogenate, liver microsome and human hepatoblastoma cell line HepG2 cells. It was found that hemin-nitrite-H2O2 could induce liver homogenate protein nitration, lipid peroxidation and liver microsome protein oxidation; it also caused a decrease of HepG2 cells viability. Baicalein, baicalin and wogonin could inhibit protein nitration and lipid peroxidation in liver homogenate as well as in HepG2 cells in a dose-dependent manner, the inhibition order was baicalein>baicalin>>wogonin. These three flavonoids also inhibited the oxidation of protein in liver microsome, the decrease of cell viability and the content of GSH in HepG2 cells, among which baicalin represented the most inhibitory effect. Besides, hemin-H2O2 induced cell injury could be augmented with the existence of nitrite, indicating protein nitration involved in hemin-nitrite-H2O2 induced liver injury. These results demonstrated hemin-nitrite-H2O2 could induce liver injury through oxidizing or nitrating different biomolecules. Baicalein, baicalin and wogonin could inhibit hemin-nitrite-H2O2 induced liver injury in dose-dependent manners by inhibiting oxidation and nitration.
Collapse
Affiliation(s)
- Yuling Zhao
- Department of Chemistry, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | | | | | | | | |
Collapse
|
25
|
Ji Y, Neverova I, Van Eyk JE, Bennett BM. Nitration of tyrosine 92 mediates the activation of rat microsomal glutathione s-transferase by peroxynitrite. J Biol Chem 2006; 281:1986-91. [PMID: 16314419 DOI: 10.1074/jbc.m509480200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
There is increasing evidence that protein function can be modified by nitration of tyrosine residue(s), a reaction catalyzed by proteins with peroxidase activity, or that occurs by interaction with peroxynitrite, a highly reactive oxidant formed by the reaction of nitric oxide with superoxide. Although there are numerous reports describing loss of function after treatment of proteins with peroxynitrite, we recently demonstrated that the microsomal glutathione S-transferase 1 is activated rather than inactivated by peroxynitrite and suggested that this could be attributed to nitration of tyrosine residues rather than to other effects of peroxynitrite. In this report, the nitrated tyrosine residues of peroxynitrite-treated microsomal glutathione S-transferase 1 were characterized by mass spectrometry and their functional significance determined. Of the seven tyrosine residues present in the protein, only those at positions 92 and 153 were nitrated after treatment with peroxynitrite. Three mutants (Y92F, Y153F, and Y92F, Y153F) were created using site-directed mutagenesis and expressed in LLC-PK1 cells. Treatment of the microsomal fractions of these cells with peroxynitrite resulted in an approximately 2-fold increase in enzyme activity in cells expressing the wild type microsomal glutathione S-transferase 1 or the Y153F mutant, whereas the enzyme activity of Y92F and double site mutant was unaffected. These results indicate that activation of microsomal glutathione S-transferase 1 by peroxynitrite is mediated by nitration of tyrosine residue 92 and represents one of the few examples in which a gain in function has been associated with nitration of a specific tyrosine residue.
Collapse
Affiliation(s)
- Yanbin Ji
- Department of Pharmacology and Toxicology, Department of Physiology, Faculty of Health Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | | | | | |
Collapse
|
26
|
Guix FX, Uribesalgo I, Coma M, Muñoz FJ. The physiology and pathophysiology of nitric oxide in the brain. Prog Neurobiol 2005; 76:126-52. [PMID: 16115721 DOI: 10.1016/j.pneurobio.2005.06.001] [Citation(s) in RCA: 496] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 06/10/2005] [Accepted: 06/14/2005] [Indexed: 12/11/2022]
Abstract
Nitric oxide (NO) is a molecule with pleiotropic effects in different tissues. NO is synthesized by NO synthases (NOS), a family with four major types: endothelial, neuronal, inducible and mitochondrial. They can be found in almost all the tissues and they can even co-exist in the same tissue. NO is a well-known vasorelaxant agent, but it works as a neurotransmitter when produced by neurons and is also involved in defense functions when it is produced by immune and glial cells. NO is thermodynamically unstable and tends to react with other molecules, resulting in the oxidation, nitrosylation or nitration of proteins, with the concomitant effects on many cellular mechanisms. NO intracellular signaling involves the activation of guanylate cyclase but it also interacts with MAPKs, apoptosis-related proteins, and mitochondrial respiratory chain or anti-proliferative molecules. It also plays a role in post-translational modification of proteins and protein degradation by the proteasome. However, under pathophysiological conditions NO has damaging effects. In disorders involving oxidative stress, such as Alzheimer's disease, stroke and Parkinson's disease, NO increases cell damage through the formation of highly reactive peroxynitrite. The paradox of beneficial and damaging effects of NO will be discussed in this review.
Collapse
Affiliation(s)
- F X Guix
- Laboratori de Fisiologia Molecular, Unitat de Senyalització Cellular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Carrer Dr. Aiguader, 80, Barcelona 08003, Spain
| | | | | | | |
Collapse
|
27
|
Shinno E, Shimoji M, Imaizumi N, Kinoshita S, Sunakawa H, Aniya Y. Activation of rat liver microsomal glutathione S-transferase by gallic acid. Life Sci 2005; 78:99-106. [PMID: 16125204 DOI: 10.1016/j.lfs.2005.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Accepted: 04/11/2005] [Indexed: 11/17/2022]
Abstract
The effect of phenolic antioxidants on the rat liver microsomal glutathione S-transferase (MGST1) was investigated in vitro. When microsomes were incubated with various polyphenolic antioxidants, gallic acid (3,4,5-trihydroxybenzoic acid) markedly increased MGST1 activity and the increase was prevented in the presence of superoxide dismutase (SOD) or catalase. The MGST1 activity increased by gallic acid was decreased by further incubation with sodium arsenite, a sulfenic acid reducing agent, but was not with dithiothreitol, a disulfide bond reducing agent. The incubation of microsomes with gallic acid in the presence of the NADPH generating system which generates reactive oxygen species (ROS) through cytochrome P-450 system increased the MGST1activity in spite of scavenging the ROS and the increase was also depressed by SOD/catalase. The increase of MGST1 activity by gallic acid was prevented by co-incubation with a stable radical, 1,1-diphenyl-2-picrylhydrazyl or ferric chloride. These results suggest that the gallic acid acts as a pro-oxidant and activates MGST1 through oxidative modification of the enzyme.
Collapse
Affiliation(s)
- Etsuki Shinno
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Moroder L. Isosteric replacement of sulfur with other chalcogens in peptides and proteins. J Pept Sci 2005; 11:187-214. [PMID: 15782428 DOI: 10.1002/psc.654] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The review addresses the functional and structural properties of the two series of chalcogen analogues of amino acids in peptides and proteins, the methionine and the serine/cysteine series, and discusses the synthesis of the related selenium/tellurium analogues as well as their use in peptide synthesis and protein expression. Advances in synthetic methodologies and recombinant technologies and their combined applications in native and expressed protein ligation allows the isomorphous character of selenium- and tellurium-containing amino acids to be exploited for production of heavy metal mutants of proteins and thus to facilitate the phasing problem in x-ray crystallography. In addition, selenocysteine has been recognized as an ideal tool for the production of selenoenzymes with new catalytic activities. Moreover, the fully isomorphous character of disulfide replacement with diselenide is well suited to increase the robustness of cystine frameworks in cystine-rich peptides and proteins and for the de novo design of even non-native cystine frameworks by exploiting the highly negative redox potential of selenols.
Collapse
Affiliation(s)
- Luis Moroder
- Max-Planck-Institut für Biochemie, D-82152 Martinsried, Germany.
| |
Collapse
|
29
|
Abstract
Microsomal glutathione transferase 1 (MGST1) is an abundant membrane-bound glutathione transferase and peroxidase constituting 3% of the endoplasmic reticulum protein in rat liver (and 5% of the outer mitochondrial membrane). The enzyme is most well studied in mammals and belongs to a large and widely distributed superfamily. Cellular and organelle protection versus oxidative stress has been demonstrated. The enzyme displays activity to a multitude of reactive substrates ranging from products of lipid peroxidation to cytostatic drugs. The methods developed for the study of MGST1 by necessity differs from that of cytosolic glutathione transferases, because detergents or lipids are included. Here, purification, assay, and preparation procedures that maintain the enzyme in its native functional state during isolation and characterization are described. Microsomal glutathione transferase 1 is activated by sulfhydryl reagents (and proteolysis), and procedures for activation and study of the activated enzyme are described. In new developments, the enzyme is studied by pre-steady state methods, as well as mass spectrometry involving direct observation of the native enzyme.
Collapse
Affiliation(s)
- Ralf Morgenstern
- Division of Biomedical Toxicology, Institute of Environmental Medicine, Karolinska Institute, Stokholm, Sweden
| |
Collapse
|
30
|
Zheng L, Settle M, Brubaker G, Schmitt D, Hazen SL, Smith JD, Kinter M. Localization of Nitration and Chlorination Sites on Apolipoprotein A-I Catalyzed by Myeloperoxidase in Human Atheroma and Associated Oxidative Impairment in ABCA1-dependent Cholesterol Efflux from Macrophages. J Biol Chem 2005; 280:38-47. [PMID: 15498770 DOI: 10.1074/jbc.m407019200] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently reported that apolipoprotein A-I (apoA-I), the major protein component of high density lipoprotein, is a selective target for myeloperoxidase (MPO)-catalyzed nitration and chlorination in both and serum of subjects with cardiovascular disease. We further showed that the extent of both apoA-I nitration and chlorination correlated with functional impairment in reverse cholesterol transport activity of the isolated lipoprotein. Herein we used tandem mass spectrometry to map the sites of MPO-mediated apoA-I nitration and chlorination in vitro and in vivo and to relate the degree of site-specific modifications to loss of apoA-I lipid binding and cholesterol efflux functions. Of the seven tyrosine residues in apoA-I, Tyr-192, Tyr-166, Tyr-236, and Tyr-29 were nitrated and chlorinated in MPO-mediated reactions. Site-specific liquid chromatography-mass spectrometry quantitative analyses demonstrated that the favored modification site following exposure to MPO-generated oxidants is Tyr-192. MPO-dependent nitration and chlorination both proceed with Tyr-166 as a secondary site and with Tyr-236 and Tyr-29 modified only minimally. Parallel functional studies demonstrated dose-dependent losses of ABCA1-dependent cholesterol acceptor and lipid binding activities with apoA-I modification by MPO. Finally tandem mass spectrometry analyses showed that apoA-I in human atherosclerotic tissue is nitrated at the MPO-preferred sites, Tyr-192 and Tyr-166. The present studies suggest that site-specific modifications of apoA-I by MPO are associated with impaired lipid binding and ABCA1-dependent cholesterol acceptor functions, providing a molecular mechanism that likely contributes to the clinical link between MPO levels and cardiovascular disease risk.
Collapse
Affiliation(s)
- Lemin Zheng
- Department of Cell Biology, Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Kinobe R, Ji Y, Nakatsu K. Peroxynitrite-mediated inactivation of heme oxygenases. BMC Pharmacol 2004; 4:26. [PMID: 15498099 PMCID: PMC529254 DOI: 10.1186/1471-2210-4-26] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Accepted: 10/21/2004] [Indexed: 11/10/2022] Open
Abstract
Background Endogenous nitric oxide (NO) and carbon monoxide (CO) are generated by nitric oxide synthase and heme oxygenase, respectively. Like NO, CO has been accepted as an important cellular signaling molecule in biological systems. An up-regulation in both gene and protein expression of heme oxygenase-1 (HO-1) under oxidative/nitrosative stress has been well documented, and the protective role of HO-1 and HO-2 against oxidative damage is proposed. However, data on the direct effect of reactive oxygen/nitrogen species (ROS/RNS) on HO function is incomplete. Using gas chromatography to quantify carbon monoxide (CO) formation from heme oxidation, we investigated the effects of peroxynitrite (ONOO-) on the in vitro catalytic activity of rat spleen (HO-1) and brain (HO-2) microsomal heme oxygenases. Results Exposure to ONOO- led to concentration-dependent but reversible decreases in the activity of microsomal rat spleen and brain HO activity. Spleen HO activity was 100-fold more sensitive to ONOO--dependent inactivation compared to that of the brain, with IC50 values of 0.015 ± 0.005 mM and 1.25 ± 0.25 mM respectively. Inhibition of both rat spleen and brain microsomal HO activity was also observed with tetra-nitromethane, a tyrosine nitrating agent, as well as two NO donors, S-nitrosoglutathione (GSNO) and diethylamine NONOate (DEA-NONOate). However, no additive effect was found following the application of NO donors and ONOO- together. Conclusion These results indicate that ONOO- may regulate HO-1 and HO-2 activities by mechanisms that involve different interactions with these proteins. It is suggested that while nitration of tyrosine residues and oxidation of sulfhydryl groups may be involved, consideration should be given to other facets of ONOO- chemistry. This inhibition of HO activity offers a mechanism for cross talk between the nitric oxide synthase and HO systems.
Collapse
Affiliation(s)
- Robert Kinobe
- Department of Pharmacology and Toxicology, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, CANADA
| | - Yanbin Ji
- Department of Pharmacology and Toxicology, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, CANADA
| | - Kanji Nakatsu
- Department of Pharmacology and Toxicology, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, CANADA
| |
Collapse
|
32
|
Rinaldi R, Aniya Y, Svensson R, Eliasson E, Swedmark S, Shimoji M, Morgenstern R. NADPH dependent activation of microsomal glutathione transferase 1. Chem Biol Interact 2004; 147:163-72. [PMID: 15013818 DOI: 10.1016/j.cbi.2003.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Revised: 12/17/2003] [Accepted: 12/17/2003] [Indexed: 10/26/2022]
Abstract
Microsomal glutathione transferase 1 (MGST1) can become activated up to 30-fold by several mechanisms in vitro (e.g. covalent modification by reactive electrophiles such as N-ethylmaleimide (NEM)). Activation has also been observed in vivo during oxidative stress. It has been noted that an NADPH generating system (g.s.) can activate MGST1 (up to 2-fold) in microsomal incubations, but the mechanism was unclear. We show here that NADPH g.s treatment impaired N-ethylmaleimide activation, indicating a shared target (identified as cysteine-49 in the latter case). Furthermore, NADPH activation was prevented by sulfhydryl compounds (glutathione and dithiothreitol). A well established candidate for activation would be oxidative stress, however we could exclude that oxidation mediated by cytochrome P450 2E1 (or flavine monooxygenase) was responsible for activation under a defined set of experimental conditions since superoxide or hydrogen peroxide alone did not activate the enzyme (in microsomes prepared by our routine procedure). Actually, the ability of MGST1 to become activated by hydrogen peroxide is critically dependent on the microsome preparation method (which influences hydrogen peroxide decomposition rate as shown here), explaining variable results in the literature. NADPH g.s. dependent activation of MGST1 could instead be explained, at least partly, by a direct effect observed also with purified enzyme (up to 1.4-fold activation). This activation was inhibited by sulfhydryl compounds and thus displays the same characteristics as that of the microsomal system. Whereas NADPH, and also ATP, activated purified MGST1, several nucleotide analogues did not, demonstrating specificity. It is thus an intriguing possibility that MGST1 function could be modulated by ligands (as well as reactive oxygen species) during oxidative stress when sulfhydryls are depleted.
Collapse
Affiliation(s)
- Rosanna Rinaldi
- Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Nobels Vg 13, Box 210, S-171 77 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
33
|
Morot-Gaudry-Talarmain Y, Rezaei H, Guermonprez L, Treguer E, Grosclaude J. Selective prion protein binding to synaptic components is modulated by oxidative and nitrosative changes induced by copper(II) and peroxynitrite in cholinergic synaptosomes, unveiling a role for calcineurin B and thioredoxin. J Neurochem 2003; 87:1456-70. [PMID: 14713301 DOI: 10.1046/j.1471-4159.2003.02111.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Choline acetyltransferase (ChAT) and choline transport are decreased after nitrosative stress. ChAT activity is altered in scrapie-infected neurons, where oxidative stress develops. Cellular prion protein (PrPc) may play a neuroprotective function in participating in the redox control of neuronal environment and regulation of copper metabolism, a role impaired when PrPc is transformed into PrPSc in prion pathologies. The complex cross-talk between PrPc and cholinergic neurons was analyzed in vitro using peroxynitrite and Cu2+ treatments on nerve endings isolated from Torpedo marmorata, a model of the motoneuron pre-synaptic element. Specific interactions between solubilized synaptic components and recombinant ovine prion protein (PrPrec) could be demonstrated by Biacore technology. Peroxynitrite abolished this interaction in a concentration-dependent way and induced significant alterations of neuronal targets. Interaction was restored by prior addition of peroxynitrite trapping agents. Cu2+ (in the form of CuSO4) treatment of synaptosomes triggered a milder oxidative effect leading to a bell-shaped increase of PrPrec binding to synaptosomal components, counteracted by the natural thiol agents, glutathione and thioredoxin. Copper(II)-induced modifications of thiols in several neuronal proteins. A positive correlation was observed between PrPrec binding and immunoreactive changes for calcineurin B and its partners, suggesting a synergy between calcineurin complex and PrP for copper regulation.
Collapse
|
34
|
van Haaften RIM, Haenen GRMM, Evelo CTA, Bast A. Effect of vitamin E on glutathione-dependent enzymes. Drug Metab Rev 2003; 35:215-53. [PMID: 12959415 DOI: 10.1081/dmr-120024086] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Reactive oxygen species and various electrophiles are involved in the etiology of diseases varying from cancer to cardiovascular and pulmonary disorders. The human body is protected against damaging effects of these compounds by a wide variety of systems. An important line of defense is formed by antioxidants. Vitamin E (consisting of various forms of tocopherols and tocotrienols) is an important fat-soluble, chain-breaking antioxidant. Besides working as an antioxidant, this compound possesses other functions with possible physiological relevance. The glutathione-dependent enzymes form another line of defense. Two important enzymes in this class are the free radical reductase and glutathione S-transferases (GSTs). The GSTs are a family of phase II detoxification enzymes. They can catalyze glutathione conjugation with various electrophiles. In most cases the electrophiles are detoxified by this conjugation, but in some cases the electrophiles are activated. Antioxidants do not act in isolation but form an intricate network. It is, for instance, known that vitamin E, together with glutathione (GSH) and a membrane-bound heat labile GSH-dependent factor, presumably an enzyme, can prevent damaging effects of reactive oxygen species on polyunsaturated fatty acids in biomembranes (lipid peroxidation). This manuscript reviews the interaction between the two defense systems, vitamin E and glutathione-dependent enzymes. On the simplest level, antioxidants such as vitamin E have protective effects on glutathione-dependent enzymes; however, we will see that reality is somewhat more complicated.
Collapse
Affiliation(s)
- Rachel I M van Haaften
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universiteit Maastricht, Maastricht, The Netherlands.
| | | | | | | |
Collapse
|