51
|
Glutathione "Redox Homeostasis" and Its Relation to Cardiovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5028181. [PMID: 31210841 PMCID: PMC6532282 DOI: 10.1155/2019/5028181] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
More people die from cardiovascular diseases (CVD) than from any other cause. Cardiovascular complications are thought to arise from enhanced levels of free radicals causing impaired "redox homeostasis," which represents the interplay between oxidative stress (OS) and reductive stress (RS). In this review, we compile several experimental research findings that show sustained shifts towards OS will alter the homeostatic redox mechanism to cause cardiovascular complications, as well as findings that show a prolonged antioxidant state or RS can similarly lead to such cardiovascular complications. This experimental evidence is specifically focused on the role of glutathione, the most abundant antioxidant in the heart, in a redox homeostatic mechanism that has been shifted towards OS or RS. This may lead to impairment of cellular signaling mechanisms and elevated pools of proteotoxicity associated with cardiac dysfunction.
Collapse
|
52
|
Guerby P, Swiader A, Augé N, Parant O, Vayssière C, Uchida K, Salvayre R, Negre-Salvayre A. High glutathionylation of placental endothelial nitric oxide synthase in preeclampsia. Redox Biol 2019; 22:101126. [PMID: 30738311 PMCID: PMC6370867 DOI: 10.1016/j.redox.2019.101126] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/25/2019] [Indexed: 12/14/2022] Open
Abstract
Decreased nitric oxide (NO) bioavailability plays a critical role in the pathophysiology of preeclampsia (PE). Recent evidence indicates that S-glutathionylation may occur on the endothelial nitric oxide synthase (eNOS), leading to eNOS uncoupling, characterized by a decreased NO production and an increased generation of superoxide anion (O2•-). We hypothesized that eNOS glutathionylation may occur in PE placentas and participate in eNOS dysfunction. The glutathionylation of eNOS was investigated in thirteen PE-affected patients and in nine normal pregnancies. Immunofluorescence, confocal microscopy and western-blot experiments carried out on eNOS immunoprecipitates, revealed a high level of eNOS glutathionylation in PE placentas, mostly reversed by dithiotreitol (DTT), thus indicative of S-glutathionylation. In order to investigate whether eNOS glutathionylation may alter trophoblast migration, an important event occurring during early placentation, cultured HTR-8/SVneo human trophoblasts (HTR8) were exposed either to low pO2 (O2 1%) or to pO2 changes (O2 1-20%), in order to generate oxidative stress. Trophoblasts exposed to low pO2, did not undergo oxidative stress nor eNOS S-glutathionylation, and were able to generate NO and migrate in a wound closure model. In contrast, trophoblasts submitted to low/high pO2 changes, exhibited oxidative stress and a (DTT reversible) S-glutathionylation of eNOS, associated with reduced NO production and migration. The autonomous production of NO seemed necessary for the migratory potential of HTR8, as suggested by the inhibitory effect of eNOS silencing by small interfering RNAs, and the eNOS inhibitor L-NAME, in low pO2 conditions. Finally, the addition of the NO donor, NOC-18 (5 µM), restored in part the migration of HTR8, thereby emphasizing the role of NO in trophoblast homeostasis. In conclusion, the high level of eNOS S-glutathionylation in PE placentas provides new insights in the mechanism of eNOS dysfunction in this disease.
Collapse
Affiliation(s)
- Paul Guerby
- Inserm U-1048, Université de Toulouse, France; Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | | | | | - Olivier Parant
- Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | - Christophe Vayssière
- Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | - Koji Uchida
- Laboratory of Food Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Japan
| | | | | |
Collapse
|
53
|
Kaschula CH, Tuveri R, Ngarande E, Dzobo K, Barnett C, Kusza DA, Graham LM, Katz AA, Rafudeen MS, Parker MI, Hunter R, Schäfer G. The garlic compound ajoene covalently binds vimentin, disrupts the vimentin network and exerts anti-metastatic activity in cancer cells. BMC Cancer 2019; 19:248. [PMID: 30894168 PMCID: PMC6425727 DOI: 10.1186/s12885-019-5388-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/20/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Garlic has been used for centuries for its flavour and health promoting properties that include protection against cancer. The vinyl disulfide-sulfoxide ajoene is one of the phytochemicals found in crushed cloves, hypothesised to act by S-thiolating reactive cysteines in target proteins. METHODS Using our fluorescently labelled ajoene analogue called dansyl-ajoene, ajoene's protein targets in MDA-MB-231 breast cancer cells were tagged and separated by 2D electrophoresis. A predominant band was identified by MALDI-TOF MS/MS to be vimentin. Target validation experiments were performed using pure recombinant vimentin protein. Computational modelling of vimentin bound to ajoene was performed using Schrödinger and pKa calculations by Epik software. Cytotoxicity of ajoene in MDA-MB-231 and HeLa cells was measured by the MTT assay. The vimentin filament network was visualised in ajoene-treated and non-treated cells by immunofluorescence and vimentin protein expression was determined by immunoblot. The invasion and migration activity was measured by wound healing and transwell assays using wildtype cells and cells in which the vimentin protein had been transiently knocked down by siRNA or overexpressed. RESULTS The dominant protein tagged by dansyl-ajoene was identified to be the 57 kDa protein vimentin. The vimentin target was validated to reveal that ajoene and dansyl-ajoene covalently bind to recombinant vimentin via a disulfide linkage at Cys-328. Computational modelling showed Cys-328 to be exposed at the termini of the vimentin tetramer. Treatment of MDA-MB-231 or HeLa cells with a non-cytotoxic concentration of ajoene caused the vimentin filament network to condense; and to increase vimentin protein expression. Ajoene inhibited the invasion and migration of both cancer cell lines which was found to be dependent on the presence of vimentin. Vimentin overexpression caused cells to become more migratory, an effect that was completely rescued by ajoene. CONCLUSIONS The garlic-derived phytochemical ajoene targets and covalently modifies vimentin in cancer cells by S-thiolating Cys-328. This interaction results in the disruption of the vimentin filament network and contributes to the anti-metastatic activity of ajoene in cancer cells.
Collapse
Affiliation(s)
- Catherine H. Kaschula
- Department of Chemistry and Polymer Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| | - Rosanna Tuveri
- Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Italy
| | - Ellen Ngarande
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Kevin Dzobo
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), UCT Medical Campus, Anzio Rd, Observatory, Cape Town, 7925 South Africa
| | - Christopher Barnett
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Daniel A. Kusza
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Lisa M. Graham
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Arieh A. Katz
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Mohamed Suhail Rafudeen
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - M. Iqbal Parker
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Roger Hunter
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Georgia Schäfer
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| |
Collapse
|
54
|
Scirè A, Cianfruglia L, Minnelli C, Bartolini D, Torquato P, Principato G, Galli F, Armeni T. Glutathione compartmentalization and its role in glutathionylation and other regulatory processes of cellular pathways. Biofactors 2019; 45:152-168. [PMID: 30561781 DOI: 10.1002/biof.1476] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
Glutathione is considered the major non-protein low molecular weight modulator of redox processes and the most important thiol reducing agent of the cell. The biosynthesis of glutathione occurs in the cytosol from its constituent amino acids, but this tripeptide is also present in the most important cellular districts, such as mitochondria, nucleus, and endoplasmic reticulum, thus playing a central role in several metabolic pathways and cytoprotection mechanisms. Indeed, glutathione is involved in the modulation of various cellular processes and, not by chance, it is a ubiquitous determinant for redox signaling, xenobiotic detoxification, and regulation of cell cycle and death programs. The balance between its concentration and redox state is due to a complex series of interactions between biosynthesis, utilization, degradation, and transport. All these factors are of great importance to understand the significance of cellular redox balance and its relationship with physiological responses and pathological conditions. The purpose of this review is to give an overview on glutathione cellular compartmentalization. Information on its subcellular distribution provides a deeper understanding of glutathione-dependent processes and reflects the importance of compartmentalization in the regulation of specific cellular pathways. © 2018 BioFactors, 45(2):152-168, 2019.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Minnelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Desirée Bartolini
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Galli
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
55
|
Young A, Gill R, Mailloux RJ. Protein S-glutathionylation: The linchpin for the transmission of regulatory information on redox buffering capacity in mitochondria. Chem Biol Interact 2018; 299:151-162. [PMID: 30537466 DOI: 10.1016/j.cbi.2018.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/08/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023]
Abstract
Protein S-glutathionylation reactions are a ubiquitous oxidative modification required to control protein function in response to changes in redox buffering capacity. These reactions are rapid and reversible and are, for the most part, enzymatically mediated by glutaredoxins (GRX) and glutathione S-transferases (GST). Protein S-glutathionylation has been found to control a range of cell functions in response to different physiological cues. Although these reactions occur throughout the cell, mitochondrial proteins seem to be highly susceptible to reversible S-glutathionylation, a feature attributed to the unique physical properties of this organelle. Indeed, mitochondria contain a number of S-glutathionylation targets which includes proteins involved in energy metabolism, solute transport, reactive oxygen species (ROS) production, proton leaks, apoptosis, antioxidant defense, and mitochondrial fission and fusion. Moreover, it has been found that conjugation and removal of glutathione from proteins in mitochondria fulfills a number of important physiological roles and defects in these reactions can have some dire pathological consequences. Here, we provide an updated overview on mitochondrial protein S-glutathionylation reactions and their importance in cell functions and physiology.
Collapse
Affiliation(s)
- Adrian Young
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Robert Gill
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ryan J Mailloux
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
56
|
Beiraghi-Toosi A, Askarian R, Sadrabadi Haghighi F, Safarian M, Kalantari F, Hashemy SI. Burn-induced Oxidative Stress and Serum Glutathione Depletion; a Cross Sectional Study. EMERGENCY (TEHRAN, IRAN) 2018; 6:e54. [PMID: 30584570 PMCID: PMC6289156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Several studies have shown the role of oxidative stress in pathophysiology of burn injuries. This study aimed to evaluate the changes of oxidant-antioxidant levels during the week following burn injuries and its correlation with grade of burn. METHODS In this prospective cross-sectional study, changes of total glutathione, reduced glutathione (GSH), oxidized GSH (GSSG), GSH/GSSG ratio, as well as Pro-oxidant-antioxidant balance (PAB) were investigated on the 1st, 2nd and 7th days of admission in patients with > 15 % burns. RESULTS 40 patients with the mean age of 21.1 ± 14.5 were studied (47.5% male). More than 50% of patients were in the 18 - 55 years age range and over 70% had 20% - 60% grade of burn. Total serum glutathione level and GSH had significant decreasing trends (P < 0.001) and GSSG and GSH/GSSG ratio had increasing trends (p < 0.001). No significant correlation was observed between serum GSH level and the total body surface area (TBSA) of burn injury (r = 0.047; p = 0.779). The evaluation of PAB and its correlation with TBSA showed a significant and direct association between them on the 1st (coefficient = 0.516; p = 0.001), 2nd (coefficient = 0.62; p <0.001), and 3rd (coefficient = 0.471; p = 0.002) day of follow up. CONCLUSION According to this study, the redox perturbation occurred in burn injury which was measured and proved by decreased GSH/GSSG ratio as well as the shift of PAB in favour of oxidants. Besides, since PAB positively correlated with the severity of dermal damage, it might suggest the application of antioxidants as a part of therapeutic protocol for which the dosage should be proportionate to the surface area of the damaged skin.
Collapse
Affiliation(s)
- Arash Beiraghi-Toosi
- Surgical Oncology Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Plastic Surgery, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roya Askarian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Sadrabadi Haghighi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Safarian
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Kalantari
- Surgical Oncology Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
57
|
Teskey G, Abrahem R, Cao R, Gyurjian K, Islamoglu H, Lucero M, Martinez A, Paredes E, Salaiz O, Robinson B, Venketaraman V. Glutathione as a Marker for Human Disease. Adv Clin Chem 2018; 87:141-159. [PMID: 30342710 DOI: 10.1016/bs.acc.2018.07.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glutathione (GSH), often referred to as "the master antioxidant," participates not only in antioxidant defense systems, but many metabolic processes, and therefore its role cannot be overstated. GSH deficiency causes cellular risk for oxidative damage and thus as expected, GSH imbalance is observed in a wide range of pathological conditions including tuberculosis (TB), HIV, diabetes, cancer, and aging. Consequently, it is not surprising that GSH has attracted the attention of biological researchers and pharmacologists alike as a possible target for medical intervention. Here, we discuss the role GSH plays amongst these pathological conditions to illuminate how it can be used as a marker for human disease.
Collapse
Affiliation(s)
- Garrett Teskey
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Rachel Abrahem
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Ruoqiong Cao
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; College of life Sciences, Hebei University, Baoding, China
| | - Karo Gyurjian
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Hicret Islamoglu
- Department of Biological Sciences, California State Polytechnic University, Pomona, CA, United States
| | - Mariana Lucero
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Andrew Martinez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Erik Paredes
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Oscar Salaiz
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Brittanie Robinson
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Vishwanath Venketaraman
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States; Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
58
|
Protective Role of Carbonic Anhydrases III and VII in Cellular Defense Mechanisms upon Redox Unbalance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2018306. [PMID: 30154947 PMCID: PMC6098850 DOI: 10.1155/2018/2018306] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/24/2018] [Indexed: 01/07/2023]
Abstract
Under oxidative stress conditions, several constitutive cellular defense systems are activated, which involve both enzymatic systems and molecules with antioxidant properties such as glutathione and vitamins. In addition, proteins containing reactive sulfhydryl groups may eventually undergo reversible redox modifications whose products act as protective shields able to avoid further permanent molecular oxidative damage either in stressful conditions or under pathological circumstances. After the recovery of normal redox conditions, the reduced state of protein sulfhydryl groups is restored. In this context, carbonic anhydrases (CAs) III and VII, which are human metalloenzymes catalyzing the reversible hydration of carbon dioxide to bicarbonate and proton, have been identified to play an antioxidant role in cells where oxidative damage occurs. Both proteins are mainly localized in tissues characterized by a high rate of oxygen consumption, and contain on their molecular surface two reactive cysteine residues eventually undergoing S-glutathionylation. Here, we will provide an overview on the molecular and functional features of these proteins highlighting their implications into molecular processes occurring during oxidative stress conditions.
Collapse
|
59
|
Prasai PK, Shrestha B, Orr AW, Pattillo CB. Decreases in GSH:GSSG activate vascular endothelial growth factor receptor 2 (VEGFR2) in human aortic endothelial cells. Redox Biol 2018; 19:22-27. [PMID: 30096614 PMCID: PMC6086407 DOI: 10.1016/j.redox.2018.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/09/2018] [Accepted: 07/19/2018] [Indexed: 02/07/2023] Open
Abstract
The angiogenic capacity of local tissue critically regulates the response to ischemic injury. Elevated reactive oxygen species production, commonly associated with ischemic injury, has been shown to promote phosphorylation of the vascular endothelial growth factor receptor 2 (VEGFR2), a critical regulator of angiogenesis. Previous data from our lab demonstrated that diminished levels of the antioxidant glutathione positively augment ischemic angiogenesis. Here, we sought to determine the relationship between glutathione levels and oxidative stress in VEGFR2 signaling. We reveal that decreasing the ratio of GSH to GSSG with diamide leads to enhanced protein S-glutathionylation, increased reactive oxygen species (ROS) production, and enhanced VEGFR2 activation. However, increasing ROS alone was insufficient in activating VEGFR2, while ROS enhanced VEGF-stimulated VEGFR2 activation at supraphysiological levels. We also found that inhibiting glutathione reductase activity is sufficient to increase VEGFR2 activation and sensitizes cells to ROS-dependent VEGFR2 activation. Taken together, these data suggest that regulation of the cellular GSH:GSSG ratio critically regulates VEGFR2 activation. This work represents an important first step in separating thiol mediated signaling events from ROS dependent signaling.
Collapse
Affiliation(s)
- Priya K Prasai
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Bandana Shrestha
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA; Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| |
Collapse
|
60
|
Static Magnetic Fields Modulate the Response of Different Oxidative Stress Markers in a Restraint Stress Model Animal. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3960408. [PMID: 29888261 PMCID: PMC5977024 DOI: 10.1155/2018/3960408] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/29/2018] [Indexed: 12/13/2022]
Abstract
Stress is a state of vulnerable homeostasis that alters the physiological and behavioral responses. Stress induces oxidative damage in several organs including the brain, liver, kidney, stomach, and heart. Preliminary findings suggested that the magnetic stimulation could accelerate the healing processes and has been an effective complementary therapy in different pathologies. However, the mechanism of action of static magnetic fields (SMFs) is not well understood. In this study, we demonstrated the effects of static magnetic fields (0.8 mT) in a restraint stressed animal model, focusing on changes in different markers of oxidative damage. A significant increase in the plasma levels of nitric oxide (NO), malondialdehyde (MDA), and advanced oxidation protein products (AOPP), and a decrease in superoxide dismutase (SOD), glutathione (GSH), and glycation end products (AGEs) were observed in restraint stress model. Exposure to SMFs over 5 days (30, 60, and 240 min/day) caused a decrease in the NO, MDA, AGEs, and AOPP levels; in contrast, the SOD and GSH levels increased. The response to SMFs was time-dependent. Thus, we proposed that exposure to weak-intensity SMFs could offer a complementary therapy by attenuating oxidative stress. Our results provided a new perspective in health studies, particularly in the context of oxidative stress.
Collapse
|
61
|
Gaucher C, Boudier A, Bonetti J, Clarot I, Leroy P, Parent M. Glutathione: Antioxidant Properties Dedicated to Nanotechnologies. Antioxidants (Basel) 2018; 7:E62. [PMID: 29702624 PMCID: PMC5981248 DOI: 10.3390/antiox7050062] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/20/2018] [Accepted: 04/25/2018] [Indexed: 02/06/2023] Open
Abstract
Which scientist has never heard of glutathione (GSH)? This well-known low-molecular-weight tripeptide is perhaps the most famous natural antioxidant. However, the interest in GSH should not be restricted to its redox properties. This multidisciplinary review aims to bring out some lesser-known aspects of GSH, for example, as an emerging tool in nanotechnologies to achieve targeted drug delivery. After recalling the biochemistry of GSH, including its metabolism pathways and redox properties, its involvement in cellular redox homeostasis and signaling is described. Analytical methods for the dosage and localization of GSH or glutathiolated proteins are also covered. Finally, the various therapeutic strategies to replenish GSH stocks are discussed, in parallel with its use as an addressing molecule in drug delivery.
Collapse
Affiliation(s)
| | - Ariane Boudier
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France.
| | | | - Igor Clarot
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France.
| | - Pierre Leroy
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France.
| | | |
Collapse
|
62
|
Zhang X, Liu P, Zhang C, Chiewchengchol D, Zhao F, Yu H, Li J, Kambara H, Luo KY, Venkataraman A, Zhou Z, Zhou W, Zhu H, Zhao L, Sakai J, Chen Y, Ho YS, Bajrami B, Xu B, Silberstein LE, Cheng T, Xu Y, Ke Y, Luo HR. Positive Regulation of Interleukin-1β Bioactivity by Physiological ROS-Mediated Cysteine S-Glutathionylation. Cell Rep 2018; 20:224-235. [PMID: 28683316 DOI: 10.1016/j.celrep.2017.05.070] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/18/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen species (ROS)-induced cysteine S-glutathionylation is an important posttranslational modification (PTM) that controls a wide range of intracellular protein activities. However, whether physiological ROS can modulate the function of extracellular components via S-glutathionylation is unknown. Using a screening approach, we identified ROS-mediated cysteine S-glutathionylation on several extracellular cytokines. Glutathionylation of the highly conserved Cys-188 in IL-1β positively regulates its bioactivity by preventing its ROS-induced irreversible oxidation, including sulfinic acid and sulfonic acid formation. We show this mechanism protects IL-1β from deactivation by ROS in an in vivo system of irradiation-induced bone marrow (BM) injury. Glutaredoxin 1 (Grx1), an enzyme that catalyzes deglutathionylation, was present and active in the extracellular space in serum and the BM, physiologically regulating IL-1β glutathionylation and bioactivity. Collectively, we identify cysteine S-glutathionylation as a cytokine regulatory mechanism that could be a therapeutic target in the treatment of various infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Pathology and Pathophysiology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA.
| | - Peng Liu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Christie Zhang
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Direkrit Chiewchengchol
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Fan Zhao
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Hongbo Yu
- Hematopathology, Flow Cytometry, Hematology, and Blood Bank Labs, VA Boston Healthcare System, West Roxbury, MA 02132, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 01605, USA
| | - Jingyu Li
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Hiroto Kambara
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Kate Y Luo
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Arvind Venkataraman
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Ziling Zhou
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Haiyan Zhu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Li Zhao
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Jiro Sakai
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Yuanyuan Chen
- Department of Pathology and Pathophysiology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences and Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, MI 48201, USA
| | - Besnik Bajrami
- Mass Spectrometry Unit, Waters Corporation, Milford, MA 01757, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street MS015, Waltham, MA 02454, USA
| | - Leslie E Silberstein
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Tao Cheng
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yuanfu Xu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yuehai Ke
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Hongbo R Luo
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA.
| |
Collapse
|
63
|
Gill RM, O’Brien M, Young A, Gardiner D, Mailloux RJ. Protein S-glutathionylation lowers superoxide/hydrogen peroxide release from skeletal muscle mitochondria through modification of complex I and inhibition of pyruvate uptake. PLoS One 2018; 13:e0192801. [PMID: 29444156 PMCID: PMC5812644 DOI: 10.1371/journal.pone.0192801] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/30/2018] [Indexed: 01/23/2023] Open
Abstract
Protein S-glutathionylation is a reversible redox modification that regulates mitochondrial metabolism and reactive oxygen species (ROS) production in liver and cardiac tissue. However, whether or not it controls ROS release from skeletal muscle mitochondria has not been explored. In the present study, we examined if chemically-induced protein S-glutathionylation could alter superoxide (O2●-)/hydrogen peroxide (H2O2) release from isolated muscle mitochondria. Disulfiram, a powerful chemical S-glutathionylation catalyst, was used to S-glutathionylate mitochondrial proteins and ascertain if it can alter ROS production. It was found that O2●-/H2O2 release rates from permeabilized muscle mitochondria decreased with increasing doses of disulfiram (100–500 μM). This effect was highest in mitochondria oxidizing succinate or palmitoyl-carnitine, where a ~80–90% decrease in the rate of ROS release was observed. Similar effects were detected in intact mitochondria respiring under state 4 conditions. Incubation of disulfiram-treated mitochondria with DTT (2 mM) restored ROS release confirming that these effects were associated with protein S-glutathionylation. Disulfiram treatment also inhibited phosphorylating and proton leak-dependent respiration. Radiolabelled substrate uptake experiments demonstrated that disulfiram inhibited pyruvate import but had no effect on carnitine uptake. Immunoblot analysis of complex I revealed that it contained several protein S-glutathionylation targets including NDUSF1, a subunit required for NADH oxidation. Taken together, these results demonstrate that O2●-/H2O2 release from muscle mitochondria can be altered by protein S-glutathionylation. We attribute these changes to the protein S-glutathionylation complex I and inhibition of mitochondrial pyruvate carrier.
Collapse
Affiliation(s)
- Robert M. Gill
- Memorial University of Newfoundland, Department of Biochemistry, St. John’s, Newfoundland, Canada
| | - Marisa O’Brien
- Memorial University of Newfoundland, Department of Biochemistry, St. John’s, Newfoundland, Canada
| | - Adrian Young
- Memorial University of Newfoundland, Department of Biochemistry, St. John’s, Newfoundland, Canada
| | - Danielle Gardiner
- Memorial University of Newfoundland, Department of Biochemistry, St. John’s, Newfoundland, Canada
| | - Ryan J. Mailloux
- Memorial University of Newfoundland, Department of Biochemistry, St. John’s, Newfoundland, Canada
- * E-mail:
| |
Collapse
|
64
|
Shanmugam G, Narasimhan M, Tamowski S, Darley-Usmar V, Rajasekaran NS. Constitutive activation of Nrf2 induces a stable reductive state in the mouse myocardium. Redox Biol 2017; 12:937-945. [PMID: 28482326 PMCID: PMC5423345 DOI: 10.1016/j.redox.2017.04.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/26/2017] [Accepted: 04/30/2017] [Indexed: 02/01/2023] Open
Abstract
Redox homeostasis regulates key cellular signaling pathways in both physiology and pathology. The cell's antioxidant response provides a defense against oxidative stress and establishes a redox tone permissive for cell signaling. The molecular regulation of the well-known Keap1/Nrf2 system acts as sensor responding to changes in redox homeostasis and is poorly studied in the heart. Importantly, it is not yet known whether Nrf2 alone can serve as a master regulator of cellular redox homeostasis without compensation of the transcriptional regulation of antioxidant response element (ARE) genes through alternate mechanisms. Here, we addressed this question using cardiac-specific transgenic expression at two different levels of constitutively active nuclear erythroid related factor 2 (caNrf2) functioning independently of Keap1. The caNrf2 mice showed augmentation of glutathione (GSH), the key regulator of the cellular thiol redox state. The Trans-AM assay for Nrf2-binding to the antioxidant response element (ARE) showed a dose-dependent increase associated with upregulation of several major antioxidant genes and proteins. This was accompanied by a significant decrease in dihydroethidium staining and malondialdehyde (MDA) in the caNrf2-TG mice myocardium. Interestingly, caNrf2 gene-dosage dependent redox changes were noted resulting in generation of a multi-stage model of pro-reductive and reductive conditions in the myocardium of TG-low and TG-high mice, respectively. These data clearly show that Nrf2 levels alone are capable of serving as the master regulator of the ARE. These models provide an important platform to investigate the impact of the Nrf2 system independent of the need to regulate the activity of Keap1 and the consequent exposure to pro-oxidants or electrophiles, which have numerous off-target effects.
Collapse
Affiliation(s)
- Gobinath Shanmugam
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| | - Susan Tamowski
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, United States; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
65
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
66
|
O'Brien M, Chalker J, Slade L, Gardiner D, Mailloux RJ. Protein S-glutathionylation alters superoxide/hydrogen peroxide emission from pyruvate dehydrogenase complex. Free Radic Biol Med 2017; 106:302-314. [PMID: 28242228 DOI: 10.1016/j.freeradbiomed.2017.02.046] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/15/2017] [Accepted: 02/23/2017] [Indexed: 11/25/2022]
Abstract
Pyruvate dehydrogenase (Pdh) is a vital source of reactive oxygen species (ROS) in several different tissues. Pdh has also been suggested to serve as a mitochondrial redox sensor. Here, we report that O2•-/ H2O2 emission from pyruvate dehydrogenase (Pdh) is altered by S-glutathionylation. Glutathione disulfide (GSSG) amplified O2•-/ H2O2 production by purified Pdh during reverse electron transfer (RET) from NADH. Thiol oxidoreductase glutaredoxin-2 (Grx2) reversed these effects confirming that Pdh is a target for S-glutathionylation. S-glutathionylation had the opposite effect during forward electron transfer (FET) from pyruvate to NAD+ lowering O2•-/ H2O2 production. Immunoblotting for protein glutathione mixed disulfides (PSSG) following diamide treatment confirmed that purified Pdh can be S-glutathionylated. Similar observations were made with mouse liver mitochondria. S-glutathionylation catalysts diamide and disulfiram significantly reduced pyruvate or 2-oxoglutarate driven O2•-/ H2O2 production in liver mitochondria, results that were confirmed using various Pdh, 2-oxoglutarate dehydrogenase (Ogdh), and respiratory chain inhibitors. Immunoprecipitation of Pdh and Ogdh confirmed that either protein can be S-glutathionylated by diamide and disulfiram. Collectively, our results demonstrate that the S -glutathionylation of Pdh alters the amount of ROS formed by the enzyme complex. We also confirmed that Ogdh is controlled in a similar manner. Taken together, our results indicate that the redox sensing and ROS forming properties of Pdh and Ogdh are linked to S-glutathionylation.
Collapse
Affiliation(s)
- Marisa O'Brien
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Julia Chalker
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Liam Slade
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Danielle Gardiner
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9.
| |
Collapse
|
67
|
Role for NF-κB inflammatory signalling pathway in tenofovir disoproxil fumarate (TDF) induced renal damage in rats. Food Chem Toxicol 2017; 99:103-118. [DOI: 10.1016/j.fct.2016.11.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 01/28/2023]
|
68
|
Scian M, Guttman M, Bouldin SD, Outten CE, Atkins WM. The Myeloablative Drug Busulfan Converts Cysteine to Dehydroalanine and Lanthionine in Redoxins. Biochemistry 2016; 55:4720-30. [PMID: 27490699 DOI: 10.1021/acs.biochem.6b00622] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The myeloablative agent busulfan (1,4-butanediol dimethanesulfonate) is an old drug that is used routinely to eliminate cancerous bone marrow prior to hematopoietic stem cell transplant. The myeloablative activity and systemic toxicity of busulfan have been ascribed to its ability to cross-link DNA. In contrast, here we demonstrate that incubation of busulfan with the thiol redox proteins glutaredoxin or thioredoxin at pH 7.4 and 37 °C results in the formation of putative S-tetrahydrothiophenium adducts at their catalytic Cys residues, followed by β-elimination to yield dehydroalanine. Both proteins contain a second Cys, in their catalytic C-X-X-C motif, which reacts with the dehydroalanine, the initial Cys adduct with busulfan, or the S-tetrahydrothiophenium, to form novel intramolecular cross-links. The reactivity of the dehydroalanine (DHA) formed is further demonstrated by adduction with glutathione to yield a lanthionine and by a novel reaction with the reducing agent tris(2-carboxyethyl)phosphine (TCEP), which yields a phosphine adduct via Michael addition to the DHA. Formation of a second quaternary organophosphonium salt via nucleophilic substitution with TCEP on the initial busulfan-protein adduct or on the THT(+)-Redoxin species is also observed. These results reveal a rich potential for reactions of busulfan with proteins in vitro, and likely in vivo. It is striking that several of the chemically altered protein products retain none of the atoms of busulfan, in contrast to typical drug-protein adducts or traditional protein modification reagents. In particular, the ability of a clinically used drug to convert Cys to dehydrolanine in intact proteins, and its subsequent reaction with biological thiols, is unprecedented.
Collapse
Affiliation(s)
- Michele Scian
- Department of Medicinal Chemistry, University of Washington , Box 357610, Seattle, Washington 98195-7610, United States
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington , Box 357610, Seattle, Washington 98195-7610, United States
| | - Samantha D Bouldin
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
| | - Caryn E Outten
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington , Box 357610, Seattle, Washington 98195-7610, United States
| |
Collapse
|
69
|
Tamma G, Valenti G. Evaluating the Oxidative Stress in Renal Diseases: What Is the Role for S-Glutathionylation? Antioxid Redox Signal 2016; 25:147-64. [PMID: 26972776 DOI: 10.1089/ars.2016.6656] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) have long been considered as toxic derivatives of aerobic metabolism displaying a harmful effect to living cells. Deregulation of redox homeostasis and production of excessive free radicals may contribute to the pathogenesis of kidney diseases. In line, oxidative stress increases in patients with renal dysfunctions due to a general increase of ROS paralleled by impaired antioxidant ability. RECENT ADVANCES Emerging evidence revealed that physiologically, ROS can act as signaling molecules interplaying with several transduction pathways such as proliferation, differentiation, and apoptosis. ROS can exert signaling functions by modulating, at different layers, protein oxidation since proteins have "cysteine switches" that can be reversibly reduced or oxidized, supporting the dynamic signaling regulation function. In this scenario, S-glutathionylation is a posttranslational modification involved in oxidative cellular response. CRITICAL ISSUES Although it is widely accepted that renal dysfunctions are often associated with altered redox signaling, the relative role of S-glutathionylation on the pathogenesis of specific renal diseases remains unclear and needs further investigations. In this review, we discuss the impact of ROS in renal health and diseases and the role of selective S-glutathionylation proteins potentially relevant to renal physiology. FUTURE DIRECTIONS The paucity of studies linking the reversible protein glutathionylation with specific renal disorders remains unmet. The growing number of S-glutathionylated proteins indicates that this is a fascinating area of research. In this respect, further studies on the association of reversible glutathionylation with renal diseases, characterized by oxidative stress, may be useful to develop new pharmacological molecules targeting protein S-glutathionylation. Antioxid. Redox Signal. 25, 147-164.
Collapse
Affiliation(s)
- Grazia Tamma
- 1 Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy .,2 Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) , Rome, Italy
| | - Giovanna Valenti
- 1 Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy .,2 Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) , Rome, Italy .,3 Centro di Eccellenza di Genomica in campo Biomedico ed Agrario (CEGBA) , Bari, Italy
| |
Collapse
|
70
|
Glutathione Disulfide Liposomes - a Research Tool for the Study of Glutathione Disulfide Associated Functions and Dysfunctions. Biochem Biophys Rep 2016; 7:225-229. [PMID: 28409182 PMCID: PMC5386405 DOI: 10.1016/j.bbrep.2016.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Glutathione disulfide (GSSG) is the oxidized form of glutathione (GSH). GSH is a tripeptide present in the biological system in mM concentration and is the major antioxidant in the body. An increase in GSSG reflects an increase in intracellular oxidative stress and is associated with disease sates. The increase has also been demonstrated to lead to an increase in protein S-glutathionylation that can affect the structure and function of proteins. Protein S-glutathionylation serves as a regulatory mechanism during cellular oxidative stress. Though GSSG is commercially available, its roles in various GSSG-associated normal/abnormal physiological functions have not been fully delineated due to the reason that GSSG is not cell membrane permeable and a lack of method to specifically increase GSSG in cells. We have developed cationic liposomes that can effectively deliver GSSG into cells. Various concentrations of GSSG liposomes can be conveniently prepared. At 1 mg/mL, the GSSG liposomes effectively increased intracellular GSSG by 27.1 ± 6.9 folds (n = 3) in 4 hours and led to a significant increase in protein S-glutathionylation confirming that the increased GSSG is functionally effective. The Trypan blue assay demonstrated that GSSG liposomes were not cytotoxic; the cell viability was greater than 95% after cells were treated with the GSSG liposomes for 4 h. A stability study showed that the dry form of the GSSG liposomes were stable for at least 70 days when stored at -80 °C. Our data demonstrate that the GSSG liposomes can be a valuable tool in studying GSSG-associated physiological/pathological functions.
Collapse
|
71
|
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: 54] [Impact Index Per Article: 6.0] [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
|
72
|
Mailloux RJ, Craig Ayre D, Christian SL. Induction of mitochondrial reactive oxygen species production by GSH mediated S-glutathionylation of 2-oxoglutarate dehydrogenase. Redox Biol 2016; 8:285-97. [PMID: 26928132 PMCID: PMC4776629 DOI: 10.1016/j.redox.2016.02.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/01/2016] [Accepted: 02/07/2016] [Indexed: 12/18/2022] Open
Abstract
2-Oxoglutarate dehydrogenase (Ogdh) is an important mitochondria redox sensor that can undergo S-glutathionylation following an increase in H2O2 levels. Although S-glutathionylation is required to protect Ogdh from irreversible oxidation while simultaneously modulating its activity it remains unknown if glutathione can also modulate reactive oxygen species (ROS) production by the complex. We report that reduced (GSH) and oxidized (GSSG) glutathione control O2∙-/H2O2 formation by Ogdh through protein S-glutathionylation reactions. GSSG (1 mM) induced a modest decrease in Ogdh activity which was associated with a significant decrease in O2∙-/H2O2 formation. GSH had the opposite effect, amplifying O2∙-/H2O2 formation by Ogdh. Incubation of purified Ogdh in 2.5 mM GSH led to significant increase in O2∙-/H2O2 formation which also lowered NADH production. Inclusion of enzymatically active glutaredoxin-2 (Grx2) in reaction mixtures reversed the GSH-mediated amplification of O2∙-/H2O2 formation. Similarly pre-incubation of permeabilized liver mitochondria from mouse depleted of GSH showed an approximately ~3.5-fold increase in Ogdh-mediated O2∙-/H2O2 production that was matched by a significant decrease in NADH formation which could be reversed by Grx2. Taken together, our results demonstrate GSH and GSSG modulate ROS production by Ogdh through S-glutathionylation of different subunits. This is also the first demonstration that GSH can work in the opposite direction in mitochondria-amplifying ROS formation instead of quenching it. We propose that this regulatory mechanism is required to modulate ROS emission from Ogdh in response to variations in glutathione redox buffering capacity. ROS formation by Ogdh is controlled by glutathione. GSH amplifies ROS production by Ogdh. Ogdh is S-glutathionylated by GSH. Grx2 deglutathionylates Ogdh.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada.
| | - D Craig Ayre
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
73
|
Synthesis and Structure–Activity Relations in Allylsulfide and Isothiocyanate Compounds From Garlic and Broccoli Against In Vitro Cancer Cell Growth. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/b978-0-444-63749-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
74
|
Abstract
Transient multienzyme and/or multiprotein complexes (metabolons) direct substrates toward specific pathways and can significantly influence the metabolism of glutamate and glutamine in the brain. Glutamate is the primary excitatory neurotransmitter in brain. This neurotransmitter has essential roles in normal brain function including learning and memory. Metabolism of glutamate involves the coordinated activity of astrocytes and neurons and high affinity transporter proteins that are selectively distributed on these cells. This chapter describes known and possible metabolons that affect the metabolism of glutamate and related compounds in the brain, as well as some factors that can modulate the association and dissociation of such complexes, including protein modifications by acylation reactions (e.g., acetylation, palmitoylation, succinylation, SUMOylation, etc.) of specific residues. Development of strategies to modulate transient multienzyme and/or enzyme-protein interactions may represent a novel and promising therapeutic approach for treatment of diseases involving dysregulation of glutamate metabolism.
Collapse
|
75
|
Hong C, Seo H, Kwak M, Jeon J, Jang J, Jeong EM, Myeong J, Hwang YJ, Ha K, Kang MJ, Lee KP, Yi EC, Kim IG, Jeon JH, Ryu H, So I. Increased TRPC5 glutathionylation contributes to striatal neuron loss in Huntington's disease. Brain 2015; 138:3030-47. [PMID: 26133660 PMCID: PMC4643628 DOI: 10.1093/brain/awv188] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 12/22/2022] Open
Abstract
Aberrant glutathione or Ca(2+) homeostasis due to oxidative stress is associated with the pathogenesis of neurodegenerative disorders. The Ca(2+)-permeable transient receptor potential cation (TRPC) channel is predominantly expressed in the brain, which is sensitive to oxidative stress. However, the role of the TRPC channel in neurodegeneration is not known. Here, we report a mechanism of TRPC5 activation by oxidants and the effect of glutathionylated TRPC5 on striatal neurons in Huntington's disease. Intracellular oxidized glutathione leads to TRPC5 activation via TRPC5 S-glutathionylation at Cys176/Cys178 residues. The oxidized glutathione-activated TRPC5-like current results in a sustained increase in cytosolic Ca(2+), activated calmodulin-dependent protein kinase and the calpain-caspase pathway, ultimately inducing striatal neuronal cell death. We observed an abnormal glutathione pool indicative of an oxidized state in the striatum of Huntington's disease transgenic (YAC128) mice. Increased levels of endogenous TRPC5 S-glutathionylation were observed in the striatum in both transgenic mice and patients with Huntington's disease. Both knockdown and inhibition of TRPC5 significantly attenuated oxidation-induced striatal neuronal cell death. Moreover, a TRPC5 blocker improved rearing behaviour in Huntington's disease transgenic mice and motor behavioural symptoms in littermate control mice by increasing striatal neuron survival. Notably, low levels of TRPC1 increased the formation of TRPC5 homotetramer, a highly Ca(2+)-permeable channel, and stimulated Ca(2+)-dependent apoptosis in Huntington's disease cells (STHdh(Q111/111)). Taken together, these novel findings indicate that increased TRPC5 S-glutathionylation by oxidative stress and decreased TRPC1 expression contribute to neuronal damage in the striatum and may underlie neurodegeneration in Huntington's disease.
Collapse
Affiliation(s)
- Chansik Hong
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Hyemyung Seo
- 2 Department of Molecular and Life Sciences, Hanyang University, Ansan, 425-791, South Korea
| | - Misun Kwak
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Jeha Jeon
- 2 Department of Molecular and Life Sciences, Hanyang University, Ansan, 425-791, South Korea
| | - Jihoon Jang
- 2 Department of Molecular and Life Sciences, Hanyang University, Ansan, 425-791, South Korea
| | - Eui Man Jeong
- 3 Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Jongyun Myeong
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Yu Jin Hwang
- 4 VA Boston Healthcare System, Department of Neurology and Boston University Alzheimer's Disease Centre, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kotdaji Ha
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Min Jueng Kang
- 5 Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine or Pharmacy, Seoul, 110-799, South Korea
| | - Kyu Pil Lee
- 6 Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, 305-764, South Korea
| | - Eugene C Yi
- 5 Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine or Pharmacy, Seoul, 110-799, South Korea
| | - In-Gyu Kim
- 3 Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Ju-Hong Jeon
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Hoon Ryu
- 4 VA Boston Healthcare System, Department of Neurology and Boston University Alzheimer's Disease Centre, Boston University School of Medicine, Boston, MA 02118, USA 7 Centre for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 136-791, South Korea
| | - Insuk So
- 1 Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| |
Collapse
|
76
|
Mullen L, Seavill M, Hammouz R, Bottazzi B, Chan P, Vaudry D, Ghezzi P. Development of 'Redox Arrays' for identifying novel glutathionylated proteins in the secretome. Sci Rep 2015; 5:14630. [PMID: 26416726 PMCID: PMC4586893 DOI: 10.1038/srep14630] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 09/02/2015] [Indexed: 11/24/2022] Open
Abstract
Proteomics techniques for analysing the redox status of individual proteins in complex mixtures tend to identify the same proteins due to their high abundance. We describe here an array-based technique to identify proteins undergoing glutathionylation and apply it to the secretome and the proteome of human monocytic cells. The method is based on incorporation of biotinylated glutathione (GSH) into proteins, which can then be identified following binding to a 1000-protein antibody array. We thus identify 38 secreted and 55 intracellular glutathionylated proteins, most of which are novel candidates for glutathionylation. Two of the proteins identified in these experiments, IL-1 sRII and Lyn, were then confirmed to be susceptible to glutathionylation. Comparison of the redox array with conventional proteomic methods confirmed that the redox array is much more sensitive, and can be performed using more than 100-fold less protein than is required for methods based on mass spectrometry. The identification of novel targets of glutathionylation, particularly in the secretome where the protein concentration is much lower, shows that redox arrays can overcome some of the limitations of established redox proteomics techniques.
Collapse
Affiliation(s)
- Lisa Mullen
- Brighton and Sussex Medical School, Trafford Centre for Medical Research, Falmer, Brighton BN1 9RY UK
| | - Miles Seavill
- Brighton and Sussex Medical School, Trafford Centre for Medical Research, Falmer, Brighton BN1 9RY UK
| | - Raneem Hammouz
- Brighton and Sussex Medical School, Trafford Centre for Medical Research, Falmer, Brighton BN1 9RY UK
| | - Barbara Bottazzi
- Humanitas Clinical &Research Center Via Manzoni, 113, 20089 Rozzano, Milano, Italy
| | - Philippe Chan
- Platform in Proteomics PISSARO, Institute for Research and Innovation in Biomedicine, University of Rouen,76821 Mont-Saint-Aignan, France
| | - David Vaudry
- Platform in Proteomics PISSARO, Institute for Research and Innovation in Biomedicine, University of Rouen,76821 Mont-Saint-Aignan, France
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Trafford Centre for Medical Research, Falmer, Brighton BN1 9RY UK
| |
Collapse
|
77
|
Dubey M, Singh AK, Awasthi D, Nagarkoti S, Kumar S, Ali W, Chandra T, Kumar V, Barthwal MK, Jagavelu K, Sánchez-Gómez FJ, Lamas S, Dikshit M. L-Plastin S-glutathionylation promotes reduced binding to β-actin and affects neutrophil functions. Free Radic Biol Med 2015; 86:1-15. [PMID: 25881549 DOI: 10.1016/j.freeradbiomed.2015.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 03/11/2015] [Accepted: 04/03/2015] [Indexed: 01/16/2023]
Abstract
Posttranslational modifications (PTMs) of cytoskeleton proteins due to oxidative stress associated with several pathological conditions often lead to alterations in cell function. The current study evaluates the effect of nitric oxide (DETA-NO)-induced oxidative stress-related S-glutathionylation of cytoskeleton proteins in human PMNs. By using in vitro and genetic approaches, we showed that S-glutathionylation of L-plastin (LPL) and β-actin promotes reduced chemotaxis, polarization, bactericidal activity, and phagocytosis. We identified Cys-206, Cys-283, and Cys-460as S-thiolated residues in the β-actin-binding domain of LPL, where cys-460 had the maximum score. Site-directed mutagenesis of LPL Cys-460 further confirmed the role in the redox regulation of LPL. S-Thiolation diminished binding as well as the bundling activity of LPL. The presence of S-thiolated LPL was detected in neutrophils from both diabetic patients and db/db mice with impaired PMN functions. Thus, enhanced nitroxidative stress may results in LPL S-glutathionylation leading to impaired chemotaxis, polarization, and bactericidal activity of human PMNs, providing a mechanistic basis for their impaired functions in diabetes mellitus.
Collapse
Affiliation(s)
- Megha Dubey
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Abhishek K Singh
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Deepika Awasthi
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sheela Nagarkoti
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sachin Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children׳s Research Foundation, Cincinnati, OH 45229, USA
| | - Wahid Ali
- King George׳s Medical University, Lucknow, India
| | | | - Vikas Kumar
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences (NCBS-TIFR), Bangalore, India
| | - Manoj K Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | | | - Francisco J Sánchez-Gómez
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Campus Universidad Autónoma, Nicolás, Cabrera 1, E-28049, Madrid, Spain
| | - Santiago Lamas
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Campus Universidad Autónoma, Nicolás, Cabrera 1, E-28049, Madrid, Spain
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
78
|
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: 2.3] [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
|
79
|
Kaschula CH, Hunter R, Cotton J, Tuveri R, Ngarande E, Dzobo K, Schäfer G, Siyo V, Lang D, Kusza DA, Davies B, Katz AA, Parker MI. The garlic compound ajoene targets protein folding in the endoplasmic reticulum of cancer cells. Mol Carcinog 2015. [DOI: 10.1002/mc.22364] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Catherine H. Kaschula
- Department of Chemistry; University of Cape Town; Rondebosch, Cape Town South Africa
| | - Roger Hunter
- Department of Chemistry; University of Cape Town; Rondebosch, Cape Town South Africa
| | - Jonathan Cotton
- Department of Chemistry; University of Cape Town; Rondebosch, Cape Town South Africa
| | - Rossana Tuveri
- Department of Biomedical Science; University of Cagliari; Monserrato (CA) Italy
| | - Ellen Ngarande
- International Centre for Genetic Engineering and Biotechnology; Cape Town South Africa
| | - Kevin Dzobo
- Division of Medical Biochemistry; University of Cape Town; Cape Town South Africa
| | - Georgia Schäfer
- Division of Medical Biochemistry; University of Cape Town; Cape Town South Africa
- MRC/UCT Receptor Biology Unit, Institute of Infectious Disease and Molecular Medicine; University of Cape Town; Cape Town South Africa
| | - Vuyolwethu Siyo
- Division of Medical Biochemistry; University of Cape Town; Cape Town South Africa
| | - Dirk Lang
- Department of Human Biology, Division of Physiology; University of Cape Town; Cape Town South Africa
| | - Daniel A. Kusza
- Department of Chemistry; University of Cape Town; Rondebosch, Cape Town South Africa
| | - Bronwen Davies
- Department of Chemistry; University of Cape Town; Rondebosch, Cape Town South Africa
| | - Arieh A. Katz
- Division of Medical Biochemistry; University of Cape Town; Cape Town South Africa
- MRC/UCT Receptor Biology Unit, Institute of Infectious Disease and Molecular Medicine; University of Cape Town; Cape Town South Africa
| | - M. Iqbal Parker
- International Centre for Genetic Engineering and Biotechnology; Cape Town South Africa
- Division of Medical Biochemistry; University of Cape Town; Cape Town South Africa
| |
Collapse
|
80
|
McKenna MC, Rae CD. A new role for α-ketoglutarate dehydrogenase complex: regulating metabolism through post-translational modification of other enzymes. J Neurochem 2015; 134:3-6. [PMID: 26052752 DOI: 10.1111/jnc.13150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 04/28/2015] [Accepted: 04/28/2015] [Indexed: 12/26/2022]
Abstract
This Editorial highlights a study by Gibson et al. published in this issue of JNeurochem, in which the authors reveal a novel role for the α-ketoglutarate dehydrogenase complex (KGDHC) in post-translational modification of proteins. KGDHC may catalyze post-translational modification of itself as well as several other proteins by succinylation of lysine residues. The authors' report of an enzyme responsible for succinylation of key mitochondrial enzymes represents a major step toward our understanding of the complex functional metabolome. TCA, tricarboxylic acid; KG, α-ketoglutarate; KGDHC, α-ketoglutarate dehydrogenase complex; FUM, fumarase; MDH, malate dehydrogenase; ME, malic enzyme; GDH, glutamate dehydrogenase; AAT, aspartate aminotransferase; GS, glutamine synthetase; PAG, phosphate-activated glutaminase; SIRT3, silent information regulator 3; SIRT5, silent information regulator 5.
Collapse
Affiliation(s)
- Mary C McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Caroline D Rae
- Neuroscience Research Australia and School of Medical Sciences UNSW, Randwick, NSW, Australia
| |
Collapse
|
81
|
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: 4.5] [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
|
82
|
Townsend DM, Lushchak VI, Cooper AJL. A comparison of reversible versus irreversible protein glutathionylation. Adv Cancer Res 2015; 122:177-98. [PMID: 24974182 DOI: 10.1016/b978-0-12-420117-0.00005-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glutathionylation is generally a reversible posttranslational modification that occurs to cysteine residues that have been exposed to reactive oxygen species (P-SSG). This cyclical process can regulate various clusters of proteins, including those involved in critical cellular signaling functions. However, certain conditions can favor the formation of dehydroamino acids, such as 2,3-didehydroalanine (2,3-dehydroalanine, DHA) and 2,3-didehydrobutyrine (2,3-dehydrobutyrine), which can act as Michael acceptors. In turn, these can form Michael adducts with glutathione (GSH), resulting in the formation of a stable thioether conjugate, an irreversible process referred to as nonreducible glutathionylation. This is predicted to be prevalent in nature, particularly in more slowly turning over proteins. Such nonreducible glutathionylation can be distinguished from the more facile cycling signaling processes and is predicted to be of gerontological, toxicological, pharmacological, and oncological relevance. Here, we compare reversible and irreversible glutathionylation.
Collapse
Affiliation(s)
- Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Arthur J L Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA.
| |
Collapse
|
83
|
Rubino FM. Toxicity of Glutathione-Binding Metals: A Review of Targets and Mechanisms. TOXICS 2015; 3:20-62. [PMID: 29056650 PMCID: PMC5634692 DOI: 10.3390/toxics3010020] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/04/2014] [Accepted: 01/14/2015] [Indexed: 02/07/2023]
Abstract
Mercury, cadmium, arsenic and lead are among priority metals for toxicological studies due to the frequent human exposure and to the significant burden of disease following acute and chronic intoxication. Among their common characteristics is chemical affinity to proteins and non-protein thiols and their ability to generate cellular oxidative stress by the best-known Fenton mechanism. Their health effects are however diverse: kidney and liver damage, cancer at specific sites, irreversible neurological damages with metal-specific features. Mechanisms for the induction of oxidative stress by interaction with the cell thiolome will be presented, based on literature evidence and of experimental findings.
Collapse
Affiliation(s)
- Federico Maria Rubino
- LaTMA Laboratory for Analytical Toxicology and Metabonomics, Department of Health Sciences, Università degli Studi di Milano at "Ospedale San Paolo" v. A. di Rudinì 8, I-20142 Milano, Italy.
| |
Collapse
|
84
|
Mailloux RJ, Willmore WG. S-glutathionylation reactions in mitochondrial function and disease. Front Cell Dev Biol 2014; 2:68. [PMID: 25453035 PMCID: PMC4233936 DOI: 10.3389/fcell.2014.00068] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/31/2014] [Indexed: 01/23/2023] Open
Abstract
Mitochondria are highly efficient energy-transforming organelles that convert energy stored in nutrients into ATP. The production of ATP by mitochondria is dependent on oxidation of nutrients and coupling of exergonic electron transfer reactions to the genesis of transmembrane electrochemical potential of protons. Electrons can also prematurely “spin-off” from prosthetic groups in Krebs cycle enzymes and respiratory complexes and univalently reduce di-oxygen to generate reactive oxygen species (ROS) superoxide (O2•−) and hydrogen peroxide (H2O2), important signaling molecules that can be toxic at high concentrations. Production of ATP and ROS are intimately linked by the respiratory chain and the genesis of one or the other inherently depends on the metabolic state of mitochondria. Various control mechanisms converge on mitochondria to adjust ATP and ROS output in response to changing cellular demands. One control mechanism that has gained a high amount of attention recently is S-glutathionylation, a redox sensitive covalent modification that involves formation of a disulfide bridge between glutathione and an available protein cysteine thiol. A number of S-glutathionylation targets have been identified in mitochondria. It has also been established that S-glutathionylation reactions in mitochondria are mediated by the thiol oxidoreductase glutaredoxin-2 (Grx2). In the following review, emerging knowledge on S-glutathionylation reactions and its importance in modulating mitochondrial ATP and ROS production will be discussed. Major focus will be placed on Complex I of the respiratory chain since (1) it is a target for reversible S-glutathionylation by Grx2 and (2) deregulation of Complex I S-glutathionylation is associated with development of various disease states particularly heart disease. Other mitochondrial enzymes and how their S-glutathionylation profile is affected in different disease states will also be discussed.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biology, Faculty of Sciences, University of Ottawa Ottawa, ON, Canada
| | | |
Collapse
|
85
|
Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 2014; 224:164-75. [PMID: 25452175 DOI: 10.1016/j.cbi.2014.10.016] [Citation(s) in RCA: 986] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/13/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) initially considered as only damaging agents in living organisms further were found to play positive roles also. This paper describes ROS homeostasis, principles of their investigation and technical approaches to investigate ROS-related processes. Especial attention is paid to complications related to experimental documentation of these processes, their diversity, spatiotemporal distribution, relationships with physiological state of the organisms. Imbalance between ROS generation and elimination in favor of the first with certain consequences for cell physiology has been called "oxidative stress". Although almost 30years passed since the first definition of oxidative stress was introduced by Helmut Sies, to date we have no accepted classification of oxidative stress. In order to fill up this gape here classification of oxidative stress based on its intensity is proposed. Due to that oxidative stress may be classified as basal oxidative stress (BOS), low intensity oxidative stress (LOS), intermediate intensity oxidative stress (IOS), and high intensity oxidative stress (HOS). Another classification of potential interest may differentiate three categories such as mild oxidative stress (MOS), temperate oxidative stress (TOS), and finally severe (strong) oxidative stress (SOS). Perspective directions of investigations in the field include development of sophisticated classification of oxidative stresses, accurate identification of cellular ROS targets and their arranged responses to ROS influence, real in situ functions and operation of so-called "antioxidants", intracellular spatiotemporal distribution and effects of ROS, deciphering of molecular mechanisms responsible for cellular response to ROS attacks, and ROS involvement in realization of normal cellular functions in cellular homeostasis.
Collapse
Affiliation(s)
- Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Precarpathian National University named after Vassyl Stefanyk, 57 Shevchenko Str., Ivano-Frankivsk 76025, Ukraine.
| |
Collapse
|
86
|
Johnstone VPA, Hool LC. Glutathionylation of the L-type Ca2+ channel in oxidative stress-induced pathology of the heart. Int J Mol Sci 2014; 15:19203-25. [PMID: 25340983 PMCID: PMC4227269 DOI: 10.3390/ijms151019203] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 09/29/2014] [Accepted: 10/02/2014] [Indexed: 01/11/2023] Open
Abstract
There is mounting evidence to suggest that protein glutathionylation is a key process contributing to the development of pathology. Glutathionylation occurs as a result of posttranslational modification of a protein and involves the addition of a glutathione moiety at cysteine residues. Such modification can occur on a number of proteins, and exerts a variety of functional consequences. The L-type Ca2+ channel has been identified as a glutathionylation target that participates in the development of cardiac pathology. Ca2+ influx via the L-type Ca2+ channel increases production of mitochondrial reactive oxygen species (ROS) in cardiomyocytes during periods of oxidative stress. This induces a persistent increase in channel open probability, and the resulting constitutive increase in Ca2+ influx amplifies the cross-talk between the mitochondria and the channel. Novel strategies utilising targeted peptide delivery to uncouple mitochondrial ROS and Ca2+ flux via the L-type Ca2+ channel following ischemia-reperfusion have delivered promising results, and have proven capable of restoring appropriate mitochondrial function in myocytes and in vivo.
Collapse
Affiliation(s)
- Victoria P A Johnstone
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Crawley 6009, WA, Australia.
| | - Livia C Hool
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Crawley 6009, WA, Australia.
| |
Collapse
|
87
|
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
|
88
|
Stan MS, Memet I, Sima C, Popescu T, Teodorescu VS, Hermenean A, Dinischiotu A. Si/SiO2 quantum dots cause cytotoxicity in lung cells through redox homeostasis imbalance. Chem Biol Interact 2014; 220:102-15. [DOI: 10.1016/j.cbi.2014.06.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/05/2014] [Accepted: 06/19/2014] [Indexed: 11/16/2022]
|
89
|
Ebrahimizadeh W, Rajabibazl M. Bacteriophage vehicles for phage display: biology, mechanism, and application. Curr Microbiol 2014; 69:109-20. [PMID: 24638925 DOI: 10.1007/s00284-014-0557-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/26/2014] [Indexed: 11/29/2022]
Abstract
The phage display technique is a powerful tool for selection of various biological agents. This technique allows construction of large libraries from the antibody repertoire of different hosts and provides a fast and high-throughput selection method. Specific antibodies can be isolated based on distinctive characteristics from a library consisting of millions of members. These features made phage display technology preferred method for antibody selection and engineering. There are several phage display methods available and each has its unique merits and application. Selection of appropriate display technique requires basic knowledge of available methods and their mechanism. In this review, we describe different phage display techniques, available bacteriophage vehicles, and their mechanism.
Collapse
Affiliation(s)
- Walead Ebrahimizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran,
| | | |
Collapse
|
90
|
Khater S, Mohanty D. Genome-wide search for eliminylating domains reveals novel function for BLES03-like proteins. Genome Biol Evol 2014; 6:2017-33. [PMID: 25062915 PMCID: PMC4159009 DOI: 10.1093/gbe/evu161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Bacterial phosphothreonine lyases catalyze a novel posttranslational modification involving formation of dehydrobutyrine/dehyroalanine by β elimination of the phosphate group of phosphothreonine or phosphoserine residues in their substrate proteins. Though there is experimental evidence for presence of dehydro amino acids in human proteins, no eukaryotic homologs of these lyases have been identified as of today. A comprehensive genome-wide search for identifying phosphothreonine lyase homologs in eukaryotes was carried out. Our fold-based search revealed structural and catalytic site similarity between bacterial phosphothreonine lyases and BLES03 (basophilic leukemia-expressed protein 03), a human protein with unknown function. Ligand induced conformational changes similar to bacterial phosphothreonine lyases, and movement of crucial arginines in the loop region to the catalytic pocket upon binding of phosphothreonine-containing peptides was seen during docking and molecular dynamics studies. Genome-wide search for BLES03 homologs using sensitive profile-based methods revealed their presence not only in eukaryotic classes such as chordata and fungi but also in bacterial and archaebacterial classes. The synteny of these archaebacterial BLES03-like proteins was remarkably similar to that of type IV lantibiotic synthetases which harbor LanL-like phosphothreonine lyase domains. Hence, context-based analysis reinforced our earlier sequence/structure-based prediction of phosphothreonine lyase catalytic function for BLES03. Our in silico analysis has revealed that BLES03-like proteins with previously unknown function are novel eukaryotic phosphothreonine lyases involved in biosynthesis of dehydro amino acids, whereas their bacterial and archaebacterial counterparts might be involved in biosynthesis of natural products similar to lantibiotics.
Collapse
Affiliation(s)
- Shradha Khater
- Bioinformatics Center, National Institute of Immunology, New Delhi, India
| | - Debasisa Mohanty
- Bioinformatics Center, National Institute of Immunology, New Delhi, India
| |
Collapse
|
91
|
Bingham PM, Stuart SD, Zachar Z. Cancer Metabolism: A Nexus of Matter, Energy, and Reactive Oxygen Species. CANCER DRUG DISCOVERY AND DEVELOPMENT 2014. [DOI: 10.1007/978-1-4614-9545-1_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
92
|
Zarembinski TI, Doty NJ, Erickson IE, Srinivas R, Wirostko BM, Tew WP. Thiolated hyaluronan-based hydrogels crosslinked using oxidized glutathione: an injectable matrix designed for ophthalmic applications. Acta Biomater 2014; 10:94-103. [PMID: 24096152 DOI: 10.1016/j.actbio.2013.09.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 08/10/2013] [Accepted: 09/23/2013] [Indexed: 02/08/2023]
Abstract
Future ophthalmic therapeutics will require the sustained delivery of bioactive proteins and nucleic acid-based macromolecules and/or provide a suitable microenvironment for the localization and sustenance of reparative progenitor cells after transplantation into or onto the eye. Water-rich hydrogels are ideal vehicles for such cargo, but few have all the qualities desired for novel ophthalmic use, namely in situ gelation speed, cytocompatibility, biocompatibility and capacity to functionalize. We describe here the development of an ophthalmic-compatible crosslinking system using oxidized glutathione (GSSG), a physiologically relevant molecule with a history of safe use in humans. When GSSG is used in conjunction with an existing hyaluronate-based, in situ crosslinkable hydrogel platform, gels form in less than 5 min using the thiol-disulfide exchange reaction. This GSSG hydrogel supports the 3-D culture of adipose-derived stem cells in vitro and shows biocompatibility in preliminary intracutaneous and subconjunctival experiments in vivo. In addition, the thiol-disulfide exchange reaction can also be used in conjunction with other thiol-compatible chemistries to covalently link peptides for more complex formulations. These data suggest that this hydrogel could be well suited for local ocular delivery, focusing initially on front of the eye therapies. Subsequent uses of the hydrogel include delivery of back of the eye treatments and eventually into other soft, hyaluronan-rich tissues such as those from the liver and brain.
Collapse
|
93
|
Wu QS, Zou YN, Fathi Abd-Allah E. Mycorrhizal Association and ROS in Plants. OXIDATIVE DAMAGE TO PLANTS 2014:453-475. [DOI: 10.1016/b978-0-12-799963-0.00015-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
94
|
Mailloux RJ, Jin X, Willmore WG. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol 2013; 2:123-39. [PMID: 24455476 PMCID: PMC3895620 DOI: 10.1016/j.redox.2013.12.011] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/13/2022] Open
Abstract
Mitochondria have a myriad of essential functions including metabolism and apoptosis. These chief functions are reliant on electron transfer reactions and the production of ATP and reactive oxygen species (ROS). The production of ATP and ROS are intimately linked to the electron transport chain (ETC). Electrons from nutrients are passed through the ETC via a series of acceptor and donor molecules to the terminal electron acceptor molecular oxygen (O2) which ultimately drives the synthesis of ATP. Electron transfer through the respiratory chain and nutrient oxidation also produces ROS. At high enough concentrations ROS can activate mitochondrial apoptotic machinery which ultimately leads to cell death. However, if maintained at low enough concentrations ROS can serve as important signaling molecules. Various regulatory mechanisms converge upon mitochondria to modulate ATP synthesis and ROS production. Given that mitochondrial function depends on redox reactions, it is important to consider how redox signals modulate mitochondrial processes. Here, we provide the first comprehensive review on how redox signals mediated through cysteine oxidation, namely S-oxidation (sulfenylation, sulfinylation), S-glutathionylation, and S-nitrosylation, regulate key mitochondrial functions including nutrient oxidation, oxidative phosphorylation, ROS production, mitochondrial permeability transition (MPT), apoptosis, and mitochondrial fission and fusion. We also consider the chemistry behind these reactions and how they are modulated in mitochondria. In addition, we also discuss emerging knowledge on disorders and disease states that are associated with deregulated redox signaling in mitochondria and how mitochondria-targeted medicines can be utilized to restore mitochondrial redox signaling.
Collapse
Affiliation(s)
- Ryan J. Mailloux
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada K1S 5B6
- Toxicology Research Division, Food Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Xiaolei Jin
- Toxicology Research Division, Food Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - William G. Willmore
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada K1S 5B6
| |
Collapse
|
95
|
Uppugunduri CRS, Rezgui MA, Diaz PH, Tyagi AK, Rousseau J, Daali Y, Duval M, Bittencourt H, Krajinovic M, Ansari M. The association of cytochrome P450 genetic polymorphisms with sulfolane formation and the efficacy of a busulfan-based conditioning regimen in pediatric patients undergoing hematopoietic stem cell transplantation. THE PHARMACOGENOMICS JOURNAL 2013; 14:263-71. [PMID: 24165757 DOI: 10.1038/tpj.2013.38] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/06/2013] [Accepted: 09/09/2013] [Indexed: 11/09/2022]
Abstract
Cytochrome P450 enzymes (CYPs) and flavin-containing monooxygenases (FMOs) likely have a role in the oxidation of intermediate metabolites of busulfan (Bu). In vitro studies to investigate the involvement of these enzymes are cumbersome because of the volatile nature of the intermediate metabolite tetrahydrothiophene (THT) and the lack of sensitive quantitation methods. This study explored the association between the CYP2C9, CYP2C19, CYP2B6 and FMO3 genotypes and sulfolane (Su, a water soluble metabolite of Bu) plasma levels in children undergoing hematopoietic stem cell transplantation (HSCT). The relationship between these genotypes and the effectiveness of myeloablative conditioning was also analyzed. Sixty-six children receiving an intravenous Bu-based myeloablative conditioning regimen were genotyped for common functional variant alleles in CYP2C9 (*2 and *3), CYP2C19 (*2 and *17), FMO3 (rs2266780, rs2266782 and rs1736557) and CYP2B6 (*5 and *9). The plasma levels of Bu and its metabolite Su were measured after the ninth Bu dose in a subset of 44 patients for whom plasma samples were available. The ratio of Bu to Su was considered the metabolic ratio (MR) and was compared across the genotype groups. Higher MRs were observed in CYP2C9*2 and *3 allele carriers (mean±s.d.: 7.8±3.6 in carriers vs 4.4±2.2 in non-carriers; P=0.003). An increased incidence of graft failure was observed among patients with an MR>5 compared with those with MR values <5 (20% vs 0%; P=0.02). In contrast, a significantly higher incidence of relapse and graft failure (evaluated as event-free survival) was observed in patients with malignant disease who carried CYP2B6 alleles with reduced function on both chromosomes compared with carriers of at least one normal allele (100% vs 40%; P=0.0001). These results suggest that CYP2C9 has a role in the oxidation reactions of THT and indicate that it may be possible to predict the efficacy of Bu-based myeloablative conditioning before HSCT on the basis of CYP genotypes and Bu MRs.
Collapse
Affiliation(s)
- C R S Uppugunduri
- 1] Department of Pediatrics, Onco-Hematology unit, University Hospital of Geneva, Geneva, Switzerland [2] CANSEARCH Research Laboratory, Geneva Medical University, Geneva, Switzerland
| | - M A Rezgui
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - P H Diaz
- 1] Department of Pediatrics, Onco-Hematology unit, University Hospital of Geneva, Geneva, Switzerland [2] CANSEARCH Research Laboratory, Geneva Medical University, Geneva, Switzerland
| | - A K Tyagi
- 1] Department of Pediatrics, Onco-Hematology unit, University Hospital of Geneva, Geneva, Switzerland [2] CANSEARCH Research Laboratory, Geneva Medical University, Geneva, Switzerland
| | - J Rousseau
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Y Daali
- 1] CANSEARCH Research Laboratory, Geneva Medical University, Geneva, Switzerland [2] Department of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Geneva, Switzerland
| | - M Duval
- 1] Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada [2] Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - H Bittencourt
- 1] Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada [2] Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - M Krajinovic
- 1] Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada [2] Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada [3] Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
| | - M Ansari
- 1] Department of Pediatrics, Onco-Hematology unit, University Hospital of Geneva, Geneva, Switzerland [2] CANSEARCH Research Laboratory, Geneva Medical University, Geneva, Switzerland
| |
Collapse
|
96
|
Glutathione controls the redox state of the mitochondrial carnitine/acylcarnitine carrier Cys residues by glutathionylation. Biochim Biophys Acta Gen Subj 2013; 1830:5299-304. [PMID: 23948593 DOI: 10.1016/j.bbagen.2013.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/26/2013] [Accepted: 08/05/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND The mitochondrial carnitine/acylcarnitine carrier (CAC) is essential for cell metabolism since it catalyzes the transport of acylcarnitines into mitochondria allowing the β-oxidation of fatty acids. CAC functional and structural properties have been characterized. Cys residues which could form disulfides suggest the involvement of CAC in redox switches. METHODS The effect of GSH and GSSG on the [(3)H]-carnitine/carnitine antiport catalyzed by the CAC in proteoliposomes has been studied. The Cys residues involved in the redox switch have been identified by site-directed mutagenesis. Glutathionylated CAC has been assessed by glutathionyl-protein specific antibody. RESULTS GSH led to increase of transport activity of the CAC extracted from liver mitochondria. A similar effect was observed on the recombinant CAC. The presence of glutaredoxin-1 (Grx1) accelerated the GSH activation of the recombinant CAC. The effect was more evident at 37°C. GSSG led to transport inhibition which was reversed by dithioerythritol (DTE). The effects of GSH and GSSG were studied on CAC Cys-mutants. CAC lacking C136 and C155 was insensitive to both reagents. Mutants containing these two Cys responded as the wild-type. Anti-glutathionyl antibody revealed the formation of glutathionylated CAC. CONCLUSIONS CAC is redox-sensitive and it is regulated by the GSH/GSSG couple. C136 and C155 are responsible for the regulation which occurs through glutathionylation. GENERAL SIGNIFICANCE CAC is sensitive to the redox state of the cell switching between oxidized and reduced forms in response to variation of GSSG and GSH concentrations.
Collapse
|
97
|
Menon D, Board PG. A role for glutathione transferase Omega 1 (GSTO1-1) in the glutathionylation cycle. J Biol Chem 2013; 288:25769-25779. [PMID: 23888047 DOI: 10.1074/jbc.m113.487785] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The glutathionylation of intracellular protein thiols can protect against irreversible oxidation and can act as a redox switch regulating metabolic pathways. In this study we discovered that the Omega class glutathione transferase GSTO1-1 plays a significant role in the glutathionylation cycle. The catalytic activity of GSTO1-1 was determined in vitro by assaying the deglutathionylation of a synthetic peptide by tryptophan fluorescence quenching and in T47-D epithelial breast cancer cells by both immunoblotting and the direct determination of total glutathionylation. Mutating the active site cysteine residue (Cys-32) ablated the deglutathionylating activity of GSTO1-1. Furthermore, we demonstrate that the expression of GSTO1-1 in T47-D cells that are devoid of endogenous GSTO1-1 resulted in a 50% reduction in total glutathionylation levels. Mass spectrometry and immunoprecipitation identified β-actin as a protein that is specifically deglutathionylated by GSTO1-1 in T47-D cells. In contrast to the deglutathionylation activity, we also found that GSTO1-1 is associated with the rapid glutathionylation of cellular proteins when the cells are exposed to S-nitrosoglutathione. The common A140D genetic polymorphism in GSTO1 was found to have significant effects on the kinetics of both the deglutathionylation and glutathionylation reactions. Genetic variation in GSTO1-1 has been associated with a range of diseases, and the discovery that a frequent GSTO1-1 polymorphism affects glutathionylation cycle reactions reveals a common mechanism where it can act on multiple proteins and pathways.
Collapse
Affiliation(s)
- Deepthi Menon
- From the Department of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Canberra ACT-2600, Australia
| | - Philip G Board
- From the Department of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Canberra ACT-2600, Australia.
| |
Collapse
|
98
|
Lockwood TD. Lysosomal metal, redox and proton cycles influencing the CysHis cathepsin reaction. Metallomics 2013; 5:110-24. [PMID: 23302864 DOI: 10.1039/c2mt20156a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the 1930's pioneers discovered that maximal autolysis in tissue homogenates requires metal chelator, sulfhydryl reducing agent and acid pH. However, metals, reducing equivalents and protons (MR&P) have been overlooked as combined catalytic controls. Three categories of lysosomal machinery drive three distinguishable cycles importing and exporting MR&P. Zn(2+) preemptively inhibits CysHis catalysis under otherwise optimal protonation and reduction. Protein-bound cell Zn(2+) concentration is 200-2000 times the non-sequestered inhibitory concentration. Following autophagy, lysosomal proteolysis liberates much inhibitory Zn(2+). The vacuolar proton pump is the driving force for Zn(2+) export, as well as protonation of the peptidolytic mechanism. Other machinery of lysosomal cycles includes proton-driven Zn(2+) exporters (e.g. SLC11A1), Zn(2+) channels (e.g. TRPML-1), lysosomal thiol reductase, etc. The CysHis dyad is a sensor of the vacuolar environment of MR&P, an integrator of these simultaneous variables, and a catalytic responder. Rate-determination can shift between autophagic substrate acquisition (swallowing) and substrate degradation (digesting). Zn(2+) recycling from degraded proteins to new proteins is a fourth cycle that might pace lysosomal function under some conditions. Heritable insufficient or excess functions of CysHis cathepsins are associated with dysfunctional inflammation and immunity/auto-immunity, including diabetic pathogenesis.
Collapse
Affiliation(s)
- Thomas D Lockwood
- Dept. of Pharmacology, School of Medicine, Wright State University, Dayton, Ohio 45435, USA.
| |
Collapse
|
99
|
Abstract
SIGNIFICANCE Glutaredoxins (Grxs) are small oxidoreductases of the thioredoxin family of proteins regulating the thiol redox state of several proteins. Thereby, Grxs are key elements in redox signaling. RECENT ADVANCES Redox signaling via protein thiols depends on reversible oxidative modifications induced mainly by reactive oxygen/nitrogen species and glutathione (GSH) in form of its oxidized disulfide or S-nitroso-glutathione. Grxs contribute to redox signaling by the catalysis of glutathionylation, de-glutathionylation, as well as reduction of disulfide bridges via two distinct enzymatic mechanisms. The dithiol mechanism utilizes both active site cysteines to reduce disulfides, whereas the monothiol mechanism utilizes only the N-terminal active site cysteine for the reduction of GSH mixed disulfides. The sphere of action of Grxs continues to grow with the recent identification of novel targets. CRITICAL ISSUES Because of limited methodological tools, the identification of new substrates for oxidoreductases in general is one of the biggest challenges in this research area. FUTURE DIRECTIONS With this review, we provide a condensed summary of the current knowledge of thiol/disulfide exchange reactions catalyzed by Grxs regarding the mechanistic, structural, and functional aspects. The latter will be of high importance for future research directions, gaining novel insights into redox signaling in general, and the role of Grxs in particular.
Collapse
Affiliation(s)
- Christopher Horst Lillig
- Institut für Biochemie und Molekularbiologie, Universitätsmedizin Greifswald, Ernst Moritz Arndt-Universität Greifswald, Greifswald, Germany
| | | |
Collapse
|
100
|
Carnicer R, Crabtree MJ, Sivakumaran V, Casadei B, Kass DA. Nitric oxide synthases in heart failure. Antioxid Redox Signal 2013; 18:1078-99. [PMID: 22871241 PMCID: PMC3567782 DOI: 10.1089/ars.2012.4824] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/07/2012] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE The regulation of myocardial function by constitutive nitric oxide synthases (NOS) is important for the maintenance of myocardial Ca(2+) homeostasis, relaxation and distensibility, and protection from arrhythmia and abnormal stress stimuli. However, sustained insults such as diabetes, hypertension, hemodynamic overload, and atrial fibrillation lead to dysfunctional NOS activity with superoxide produced instead of NO and worse pathophysiology. RECENT ADVANCES Major strides in understanding the role of normal and abnormal constitutive NOS in the heart have revealed molecular targets by which NO modulates myocyte function and morphology, the role and nature of post-translational modifications of NOS, and factors controlling nitroso-redox balance. Localized and differential signaling from NOS1 (neuronal) versus NOS3 (endothelial) isoforms are being identified, as are methods to restore NOS function in heart disease. CRITICAL ISSUES Abnormal NOS signaling plays a key role in many cardiac disorders, while targeted modulation may potentially reverse this pathogenic source of oxidative stress. FUTURE DIRECTIONS Improvements in the clinical translation of potent modulators of NOS function/dysfunction may ultimately provide a powerful new treatment for many hearts diseases that are fueled by nitroso-redox imbalance.
Collapse
Affiliation(s)
- Ricardo Carnicer
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mark J. Crabtree
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Vidhya Sivakumaran
- Division of Cardiology, Department of Medicine, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | - Barbara Casadei
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David A. Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| |
Collapse
|