51
|
Loh JT, Su IH. Post-translational modification-regulated leukocyte adhesion and migration. Oncotarget 2018; 7:37347-37360. [PMID: 26993608 PMCID: PMC5095081 DOI: 10.18632/oncotarget.8135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/28/2016] [Indexed: 12/30/2022] Open
Abstract
Leukocytes undergo frequent phenotypic changes and rapidly infiltrate peripheral and lymphoid tissues in order to carry out immune responses. The recruitment of circulating leukocytes into inflamed tissues depends on integrin-mediated tethering and rolling of these cells on the vascular endothelium, followed by transmigration into the tissues. This dynamic process of migration requires the coordination of large numbers of cytosolic and transmembrane proteins whose functional activities are typically regulated by post-translational modifications (PTMs). Our recent studies have shown that the lysine methyltransferase, Ezh2, critically regulates integrin signalling and governs the adhesion dynamics of leukocytes via direct methylation of talin, a key molecule that controls these processes by linking integrins to the actin cytoskeleton. In this review, we will discuss the various modes of leukocyte migration and examine how PTMs of cytoskeletal/adhesion associated proteins play fundamental roles in the dynamic regulation of leukocyte migration. Furthermore, we will discuss molecular details of the adhesion dynamics controlled by Ezh2-mediated talin methylation and the potential implications of this novel regulatory mechanism for leukocyte migration, immune responses, and pathogenic processes, such as allergic contact dermatitis and tumorigenesis.
Collapse
Affiliation(s)
- Jia Tong Loh
- School of Biological Sciences, College of Science, Nanyang Technological University, Republic of Singapore
| | - I-Hsin Su
- School of Biological Sciences, College of Science, Nanyang Technological University, Republic of Singapore
| |
Collapse
|
52
|
Stojkov D, Amini P, Oberson K, Sokollik C, Duppenthaler A, Simon HU, Yousefi S. ROS and glutathionylation balance cytoskeletal dynamics in neutrophil extracellular trap formation. J Cell Biol 2017; 216:4073-4090. [PMID: 29150539 PMCID: PMC5716265 DOI: 10.1083/jcb.201611168] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/27/2017] [Accepted: 08/22/2017] [Indexed: 12/26/2022] Open
Abstract
Neutrophils can release their genomic DNA as extracellular traps (NETs), which ensnare bacteria and limit their replication. Stojkov et al. find that modulation of cytoskeletal dynamics by reactive oxygen species and glutathionylation controls the degranulation and release of mitochondrial DNA required for NET formation. The antimicrobial defense activity of neutrophils partly depends on their ability to form neutrophil extracellular traps (NETs), but the underlying mechanism controlling NET formation remains unclear. We demonstrate that inhibiting cytoskeletal dynamics with pharmacological agents or by genetic manipulation prevents the degranulation of neutrophils and mitochondrial DNA release required for NET formation. Wiskott-Aldrich syndrome protein–deficient neutrophils are unable to polymerize actin and exhibit a block in both degranulation and DNA release. Similarly, neutrophils with a genetic defect in NADPH oxidase fail to induce either actin and tubulin polymerization or NET formation on activation. Moreover, neutrophils deficient in glutaredoxin 1 (Grx1), an enzyme required for deglutathionylation of actin and tubulin, are unable to polymerize either cytoskeletal network and fail to degranulate or release DNA. Collectively, cytoskeletal dynamics are achieved as a balance between reactive oxygen species–regulated effects on polymerization and glutathionylation on the one hand and the Grx1-mediated deglutathionylation that is required for NET formation on the other.
Collapse
Affiliation(s)
- Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Poorya Amini
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Kevin Oberson
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Christiane Sokollik
- Unit of Pediatric Infectious Diseases, University Children's Hospital Bern, Bern, Switzerland
| | - Andrea Duppenthaler
- Unit of Pediatric Infectious Diseases, University Children's Hospital Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
53
|
Quiles JM, Narasimhan M, Mosbruger T, Shanmugam G, Crossman D, Rajasekaran NS. Identification of transcriptome signature for myocardial reductive stress. Redox Biol 2017; 13:568-580. [PMID: 28768233 PMCID: PMC5536881 DOI: 10.1016/j.redox.2017.07.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
The nuclear factor erythroid 2 like 2 (Nfe2l2/Nrf2) is a master regulator of antioxidant gene transcription. We recently identified that constitutive activation of Nrf2 (CaNrf2) caused reductive stress (RS) in the myocardium. Here we investigate how chronic Nrf2 activation alters myocardial mRNA transcriptome in the hearts of CaNrf2 transgenic (TG-low and TG-high) mice using an unbiased integrated systems approach and next generation RNA sequencing followed by qRT-PCR methods. A total of 246 and 1031 differentially expressed genes (DEGs) were identified in the heart of TGL and TGH in relation to NTG littermates at ~ 6 months of age. Notably, the expression and validation of the transcripts were gene-dosage dependent and statistically significant. Ingenuity Pathway Analysis identified enriched biological processes and canonical pathways associated with myocardial RS in the CaNrf2-TG mice. In addition, an overrepresentation of xenobiotic metabolic signaling, glutathione-mediated detoxification, unfolded protein response, and protein ubiquitination was observed. Other, non-canonical signaling pathways identified include: eNOS, integrin-linked kinase, glucocorticoid receptor, PI3/AKT, actin cytoskeleton, cardiac hypertrophy, and the endoplasmic reticulum stress response. In conclusion, this mRNA profiling identified a "biosignature" for pro-reductive (TGL) and reductive stress (TGH) that can predict the onset, rate of progression, and clinical outcome of Nrf2-dependent myocardial complications. We anticipate that this global sequencing analysis will illuminate the undesirable effect of chronic Nrf2 signaling leading to RS-mediated pathogenesis besides providing important guidance for the application of Nrf2 activation-based cytoprotective strategies.
Collapse
Affiliation(s)
- Justin M Quiles
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Timothy Mosbruger
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Gobinath Shanmugam
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David Crossman
- Heflin Center for Genomic Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
54
|
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
|
55
|
A Feed-Forward Mechanism Involving the NOX Complex and RyR-Mediated Ca2+ Release During Axonal Specification. J Neurosci 2017; 36:11107-11119. [PMID: 27798190 DOI: 10.1523/jneurosci.1455-16.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/06/2016] [Indexed: 01/16/2023] Open
Abstract
Physiological levels of ROS support neurite outgrowth and axonal specification, but the mechanisms by which ROS are able to shape neurons remain unknown. Ca2+, a broad intracellular second messenger, promotes both Rac1 activation and neurite extension. Ca2+ release from the endoplasmic reticulum, mediated by both the IP3R1 and ryanodine receptor (RyR) channels, requires physiological ROS levels that are mainly sustained by the NADPH oxidase (NOX) complex. In this work, we explore the contribution of the link between NOX and RyR-mediated Ca2+ release toward axonal specification of rat hippocampal neurons. Using genetic approaches, we find that NOX activation promotes both axonal development and Rac1 activation through a RyR-mediated mechanism, which in turn activates NOX through Rac1, one of the NOX subunits. Collectively, these data suggest a feedforward mechanism that integrates both NOX activity and RyR-mediated Ca2+ release to support cellular mechanisms involved in axon development. SIGNIFICANCE STATEMENT High levels of ROS are frequently associated with oxidative stress and disease. In contrast, physiological levels of ROS, mainly sustained by the NADPH oxidase (NOX) complex, promote neuronal development and axonal growth. However, the mechanisms by which ROS shape neurons have not been described. Our work suggests that NOX-derived ROS promote axonal growth by regulating Rac1 activity, a molecular determinant of axonal growth, through a ryanodine receptor (RyR)-mediated Ca2+ release mechanism. In addition, Rac1, one of the NOX subunits, was activated after RyR-mediated Ca2+ release, suggesting a feedforward mechanism between NOX and RyR. Collectively, our data suggest a novel mechanism that is instrumental in sustaining physiological levels of ROS required for axonal growth of hippocampal neurons.
Collapse
|
56
|
Bertling E, Hotulainen P. New waves in dendritic spine actin cytoskeleton: From branches and bundles to rings, from actin binding proteins to post-translational modifications. Mol Cell Neurosci 2017; 84:77-84. [PMID: 28479292 DOI: 10.1016/j.mcn.2017.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/24/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the number or strength of synapses are physiological mechanisms behind learning. The growth and maturation of dendritic spines and the activity-induced changes to their morphology are all based on changes to the actin cytoskeleton. In this review, we will discuss the regulation of the actin cytoskeleton in dendritic spine formation and maturation, as well as in synaptic strengthening. Concerning spine formation, we will focus on spine initiation, which has received less attention in the literature. We will also examine the recently revealed regulation of the actin cytoskeleton through post-translational modifications of actin monomers, in addition to the conventional regulation of actin via actin-binding proteins.
Collapse
Affiliation(s)
- Enni Bertling
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
57
|
Modulation of the specific glutathionylation of mitochondrial proteins in the yeast Saccharomyces cerevisiae under basal and stress conditions. Biochem J 2017; 474:1175-1193. [PMID: 28167699 DOI: 10.1042/bcj20160927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/27/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
The potential biological consequences of oxidative stress and changes in glutathione levels include the oxidation of susceptible protein thiols and reversible covalent binding of glutathione to the -SH groups of proteins by S-glutathionylation. Mitochondria are central to the response to oxidative stress and redox signaling. It is therefore crucial to explore the adaptive response to changes in thiol-dependent redox status in these organelles. We optimized the purification protocol of glutathionylated proteins in the yeast Saccharomyces cerevisiae and present a detailed proteomic analysis of the targets of protein glutathionylation in cells undergoing constitutive metabolism and after exposure to various stress conditions. This work establishes the physiological importance of the glutathionylation process in S. cerevisiae under basal conditions and provides evidence for an atypical and unexpected cellular distribution of the process between the cytosol and mitochondria. In addition, our data indicate that each oxidative condition (diamide, GSSG, H2O2, or the presence of iron) elicits an adaptive metabolic response affecting specific mitochondrial metabolic pathways, mainly involved in the energetic maintenance of the cells. The correlation of protein modifications with intracellular glutathione levels suggests that protein deglutathionylation may play a role in protecting mitochondria from oxidative stress. This work provides further insights into the diversity of proteins undergoing glutathionylation and the role of this post-translational modification as a regulatory process in the adaptive response of the cell.
Collapse
|
58
|
Ercolani L, Scirè A, Galeazzi R, Massaccesi L, Cianfruglia L, Amici A, Piva F, Urbanelli L, Emiliani C, Principato G, Armeni T. A possible S-glutathionylation of specific proteins by glyoxalase II: An in vitro and in silico study. Cell Biochem Funct 2017; 34:620-627. [PMID: 27935136 DOI: 10.1002/cbf.3236] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/14/2016] [Accepted: 10/14/2016] [Indexed: 01/07/2023]
Abstract
Glyoxalase II, the second of 2 enzymes in the glyoxalase system, is a hydroxyacylglutathione hydrolase that catalyses the hydrolysis of S-d-lactoylglutathione to form d-lactic acid and glutathione, which is released from the active site. The tripeptide glutathione is the major sulfhydryl antioxidant and has been shown to control several functions, including S-glutathionylation of proteins. S-Glutathionylation is a way for the cells to store reduced glutathione during oxidative stress, or to protect protein thiol groups from irreversible oxidation, and few enzymes involved in protein S-glutathionylation have been found to date. In this work, the enzyme glyoxalase II and its substrate S-d-lactoylglutathione were incubated with malate dehydrogenase or with actin, resulting in a glutathionylation reaction. Glyoxalase II was also submitted to docking studies. Computational data presented a high propensity of the enzyme to interact with malate dehydrogenase or actin through its catalytic site and further in silico investigation showed a high folding stability of glyoxalase II toward its own reaction product glutathione both protonated and unprotonated. This study suggests that glyoxalase II, through a specific interaction of its catalytic site with target proteins, could be able to perform a rapid and specific protein S-glutathionylation using its natural substrate S-d-lactoylglutathione. SIGNIFICANCE This article reports for the first time a possible additional role of Glo2 that, after interacting with a target protein, is able to promote S-glutathionylation using its natural substrate SLG, a glutathione derived compound. In this perspective, Glo2 can play a new important regulatory role inS-glutathionylation, acquiring further significance in cellular post-translational modifications of proteins.
Collapse
Affiliation(s)
- Luisa Ercolani
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Roberta Galeazzi
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Luca Massaccesi
- 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
| | - Adolfo Amici
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Piva
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Lorena Urbanelli
- Department of Experimental Medicine and Biochemical Sciences, Università di Perugia, Perugia, Italy
| | - Carla Emiliani
- Department of Experimental Medicine and Biochemical Sciences, Università di Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
59
|
Han J, Weisbrod RM, Shao D, Watanabe Y, Yin X, Bachschmid MM, Seta F, Janssen-Heininger YMW, Matsui R, Zang M, Hamburg NM, Cohen RA. The redox mechanism for vascular barrier dysfunction associated with metabolic disorders: Glutathionylation of Rac1 in endothelial cells. Redox Biol 2016; 9:306-319. [PMID: 27693992 PMCID: PMC5045950 DOI: 10.1016/j.redox.2016.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/08/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Oxidative stress is implicated in increased vascular permeability associated with metabolic disorders, but the underlying redox mechanism is poorly defined. S-glutathionylation, a stable adduct of glutathione with protein sulfhydryl, is a reversible oxidative modification of protein and is emerging as an important redox signaling paradigm in cardiovascular physiopathology. The present study determines the role of protein S-glutathionylation in metabolic stress-induced endothelial cell permeability. METHODS AND RESULTS In endothelial cells isolated from patients with type-2 diabetes mellitus, protein S-glutathionylation level was increased. This change was also observed in aortic endothelium in ApoE deficient (ApoE-/-) mice fed on Western diet. Metabolic stress-induced protein S-glutathionylation in human aortic endothelial cells (HAEC) was positively correlated with elevated endothelial cell permeability, as reflected by disassembly of cell-cell adherens junctions and cortical actin structures. These impairments were reversed by adenoviral overexpression of a specific de-glutathionylation enzyme, glutaredoxin-1 in cultured HAECs. Consistently, transgenic overexpression of human Glrx-1 in ApoE-/- mice fed the Western diet attenuated endothelial protein S-glutathionylation, actin cytoskeletal disorganization, and vascular permeability in the aorta. Mechanistically, glutathionylation and inactivation of Rac1, a small RhoGPase, were associated with endothelial hyperpermeability caused by metabolic stress. Glutathionylation of Rac1 on cysteine 81 and 157 located adjacent to guanine nucleotide binding site was required for the metabolic stress to inhibit Rac1 activity and promote endothelial hyperpermeability. CONCLUSIONS Glutathionylation and inactivation of Rac1 in endothelial cells represent a novel redox mechanism of vascular barrier dysfunction associated with metabolic disorders.
Collapse
Affiliation(s)
- Jingyan Han
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA.
| | - Robert M Weisbrod
- Evans Department of Medicine and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Di Shao
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Yosuke Watanabe
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoyan Yin
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Markus M Bachschmid
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Francesca Seta
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | | | - Reiko Matsui
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Mengwei Zang
- Department of Molecular Medicine, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Naomi M Hamburg
- Evans Department of Medicine and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Richard A Cohen
- Vascular Biology Section, Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
60
|
Terrill JR, Duong MN, Turner R, Le Guiner C, Boyatzis A, Kettle AJ, Grounds MD, Arthur PG. Levels of inflammation and oxidative stress, and a role for taurine in dystropathology of the Golden Retriever Muscular Dystrophy dog model for Duchenne Muscular Dystrophy. Redox Biol 2016; 9:276-286. [PMID: 27611888 PMCID: PMC5018082 DOI: 10.1016/j.redox.2016.08.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 08/24/2016] [Accepted: 08/29/2016] [Indexed: 01/29/2023] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal skeletal muscle wasting disease presenting with excessive myofibre necrosis and increased inflammation and oxidative stress. In the mdx mouse model of DMD, homeostasis of the amino acid taurine is altered, and taurine administration drastically decreases muscle necrosis, dystropathology, inflammation and protein thiol oxidation. Since the severe pathology of the Golden Retriever Muscular Dystrophy (GRMD) dog model more closely resembles the human DMD condition, we aimed to assess the generation of oxidants by inflammatory cells and taurine metabolism in this species. In muscles of 8 month GRMD dogs there was an increase in the content of neutrophils and macrophages, and an associated increase in elevated myeloperoxidase, a protein secreted by neutrophils that catalyses production of the highly reactive hypochlorous acid (HOCl). There was also increased chlorination of tyrosines, a marker of HOCl generation, increased thiol oxidation of many proteins and irreversible oxidative protein damage. Taurine, which functions as an antioxidant by trapping HOCl, was reduced in GRMD plasma; however taurine was increased in GRMD muscle tissue, potentially due to increased muscle taurine transport and synthesis. These data indicate a role for HOCl generated by neutrophils in the severe dystropathology of GRMD dogs, which may be exacerbated by decreased availability of taurine in the blood. These novel data support continued research into the precise roles of oxidative stress and taurine in DMD and emphasise the value of the GRMD dogs as a suitable pre-clinical model for testing taurine as a therapeutic intervention for DMD boys.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia, Australia.
| | - Marisa N Duong
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia, Australia
| | - Rufus Turner
- Centre for Free Radical Research, Department of Pathology, the University of Otago, Christchurch, New Zealand
| | - Caroline Le Guiner
- Atlantic Gene Therapies, INSERM UMR1089, Nantes, France; Genethon, Evry, France
| | - Amber Boyatzis
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia, Australia
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology, the University of Otago, Christchurch, New Zealand
| | - Miranda D Grounds
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia, Australia
| | - Peter G Arthur
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
61
|
Wang G, Cao L, Liu X, Sieracki NA, Di A, Wen X, Chen Y, Taylor S, Huang X, Tiruppathi C, Zhao YY, Song Y, Gao X, Jin T, Bai C, Malik AB, Xu J. Oxidant Sensing by TRPM2 Inhibits Neutrophil Migration and Mitigates Inflammation. Dev Cell 2016; 38:453-62. [PMID: 27569419 DOI: 10.1016/j.devcel.2016.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/22/2016] [Accepted: 07/18/2016] [Indexed: 02/05/2023]
Abstract
Blood neutrophils perform an essential host-defense function by directly migrating to bacterial invasion sites to kill bacteria. The mechanisms mediating the transition from the migratory to bactericidal phenotype remain elusive. Here, we demonstrate that TRPM2, a trp superfamily member, senses neutrophil-generated reactive oxygen species and restrains neutrophil migration. The inhibitory function of oxidant sensing by TRPM2 requires the oxidation of Cys549, which then induces TRMP2 binding to formyl peptide receptor 1 (FPR1) and subsequent FPR1 internalization and signaling inhibition. The oxidant sensing-induced termination of neutrophil migration at the site of infection permits a smooth transition to the subsequent microbial killing phase.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Luyang Cao
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Xiaowen Liu
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Nathan A Sieracki
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Anke Di
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Xi Wen
- Chemotaxis Signal Section, Laboratory of Immunogenetics, NIAID, NIH, Bethesda, MD 20892, USA
| | - Yong Chen
- Proteomic Core Facility, NHLBI, NIH, Bethesda, MD 20824, USA
| | - Shalina Taylor
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Xiaojia Huang
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | | | - You-Yang Zhao
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaopei Gao
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Tian Jin
- Chemotaxis Signal Section, Laboratory of Immunogenetics, NIAID, NIH, Bethesda, MD 20892, USA
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA
| | - Jingsong Xu
- Department of Pharmacology, University of Illinois, Chicago, IL 60612, USA; Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
62
|
Wilson C, Terman JR, González-Billault C, Ahmed G. Actin filaments-A target for redox regulation. Cytoskeleton (Hoboken) 2016; 73:577-595. [PMID: 27309342 DOI: 10.1002/cm.21315] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/03/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
Actin and its ability to polymerize into dynamic filaments is critical for the form and function of cells throughout the body. While multiple proteins have been characterized as affecting actin dynamics through noncovalent means, actin and its protein regulators are also susceptible to covalent modifications of their amino acid residues. In this regard, oxidation-reduction (Redox) intermediates have emerged as key modulators of the actin cytoskeleton with multiple different effects on cellular form and function. Here, we review work implicating Redox intermediates in post-translationally altering actin and discuss what is known regarding how these alterations affect the properties of actin. We also focus on two of the best characterized enzymatic sources of these Redox intermediates-the NADPH oxidase NOX and the flavoprotein monooxygenase MICAL-and detail how they have both been identified as altering actin, but share little similarity and employ different means to regulate actin dynamics. Finally, we discuss the role of these enzymes and redox signaling in regulating the actin cytoskeleton in vivo and highlight their importance for neuronal form and function in health and disease. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Carlos Wilson
- Department of Biology, Faculty of Sciences, Universidad De Chile, Las Palmeras 3425, Santiago, 7800024, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Jonathan R Terman
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas, 75390. .,Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas, 75390.
| | - Christian González-Billault
- Department of Biology, Faculty of Sciences, Universidad De Chile, Las Palmeras 3425, Santiago, 7800024, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. .,The Buck Institute for Research on Aging, Novato, California 94945.
| | - Giasuddin Ahmed
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas, 75390.,Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| |
Collapse
|
63
|
Love DT, Barrett TJ, White MY, Cordwell SJ, Davies MJ, Hawkins CL. Cellular targets of the myeloperoxidase-derived oxidant hypothiocyanous acid (HOSCN) and its role in the inhibition of glycolysis in macrophages. Free Radic Biol Med 2016; 94:88-98. [PMID: 26898502 DOI: 10.1016/j.freeradbiomed.2016.02.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 12/20/2022]
Abstract
Myeloperoxidase (MPO) released at sites of inflammation catalyzes the formation of the oxidants hypochlorous acid (HOCl) and hypothiocyanous acid (HOSCN) from H2O2 and halide and pseudo-halide ions. HOCl, a major oxidant produced under physiological conditions reacts rapidly with many biological molecules, and is strongly linked with tissue damage during inflammatory disease. The role of HOSCN in disease is less clear, though it can initiate cellular damage by pathways involving the selective oxidation of thiol-containing proteins. Utilizing a thiol-specific proteomic approach, we explored the cellular targets of HOSCN in macrophages (J774A.1). We report that multiple thiol-containing proteins involved in metabolism and glycolysis; fructose bisphosphate aldolase, triosephosphate isomerase, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and creatine kinase, together with a number of chaperone, antioxidant and structural proteins, were modified in a reversible manner in macrophages treated with HOSCN. The modification of the metabolic enzymes was associated with a decrease in basal glycolysis, glycolytic reserve, glycolytic capacity and lactate release, which was only partly reversible on further incubation in the absence of HOSCN. Inhibition of glycolysis preceded cell death and was seen in cells exposed to low concentrations (≤25µM) of HOSCN. The ability of HOSCN to inhibit glycolysis and perturb energy production is likely to contribute to the cell death seen in macrophages on further incubation after the initial treatment period, which may be relevant for the propagation of inflammatory disease in smokers, who have elevated plasma levels of the HOSCN precursor, thiocyanate.
Collapse
Affiliation(s)
- Dominic T Love
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Tessa J Barrett
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Melanie Y White
- School of Molecular Bioscience, School of Medical Sciences, and Charles Perkins Centre, University of Sydney, 2006, Australia
| | - Stuart J Cordwell
- School of Molecular Bioscience, School of Medical Sciences, and Charles Perkins Centre, University of Sydney, 2006, Australia
| | - Michael J Davies
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark
| | - Clare L Hawkins
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
64
|
Terrill JR, Pinniger GJ, Graves JA, Grounds MD, Arthur PG. Increasing taurine intake and taurine synthesis improves skeletal muscle function in the mdx mouse model for Duchenne muscular dystrophy. J Physiol 2016; 594:3095-110. [PMID: 26659826 DOI: 10.1113/jp271418] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/18/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease associated with increased inflammation, oxidative stress and myofibre necrosis. Cysteine precursor antioxidants such as N-acetyl cysteine (NAC) and l-2-oxothiazolidine-4-carboxylate (OTC) reduce dystropathology in the mdx mouse model for DMD, and we propose this is via increased synthesis of the amino acid taurine. We compared the capacity of OTC and taurine treatment to increase taurine content of mdx muscle, as well as effects on in vivo and ex vivo muscle function, inflammation and oxidative stress. Both treatments increased taurine in muscles, and improved many aspects of muscle function and reduced inflammation. Taurine treatment also reduced protein thiol oxidation and was overall more effective, as OTC treatment reduced body and muscle weight, suggesting some adverse effects of this drug. These data suggest that increasing dietary taurine is a better candidate for a therapeutic intervention for DMD. ABSTRACT Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease for which there is no widely available cure. Whilst the mechanism of loss of muscle function in DMD and the mdx mouse model are not fully understood, disruptions in intracellular calcium homeostasis, inflammation and oxidative stress are implicated. We have shown that protein thiol oxidation is increased in mdx muscle, and that the indirect thiol antioxidant l-2-oxothiazolidine-4-carboxylate (OTC), which increases cysteine availability, decreases pathology and increases in vivo strength. We propose that the protective effects of OTC are a consequence of conversion of cysteine to taurine, which has itself been shown to be beneficial to mdx pathology. This study compares the efficacy of taurine with OTC in decreasing dystropathology in mdx mice by measuring in vivo and ex vivo contractile function and measurements of inflammation and protein thiol oxidation. Increasing the taurine content of mdx muscle improved both in vivo and ex vivo muscle strength and function, potentially via anti-inflammatory and antioxidant effects of taurine. OTC treatment increased taurine synthesis in the liver and taurine content of mdx muscle, improved muscle function and decreased inflammation. However, OTC was less effective than taurine treatment, with OTC also decreasing body and EDL muscle weights, suggesting that OTC had some detrimental effects. These data support continued research into the use of taurine as a therapeutic intervention for DMD, and suggest that increasing dietary taurine is the better strategy for increasing taurine content and decreasing severity of dystropathology.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia.,School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Gavin J Pinniger
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Jamie A Graves
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Miranda D Grounds
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Peter G Arthur
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia
| |
Collapse
|
65
|
M. Bansbach H, H. Guilford W. Actin nitrosylation and its effect on myosin driven motility. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.3.426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
66
|
Development of a Clickable Probe for Profiling of Protein Glutathionylation in the Central Cellular Metabolism of E. coli and Drosophila. ACTA ACUST UNITED AC 2015; 22:1461-1469. [PMID: 26526100 DOI: 10.1016/j.chembiol.2015.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 11/24/2022]
Abstract
Protein glutathionylation is an important post-translational modification that regulates many cellular processes, including energy metabolism, signal transduction, and protein homeostasis. Global profiling of glutathionylated proteins (denoted as glutathionylome) is crucial for understanding redox-regulated signal transduction. Here, we developed a novel method based on click reaction and proteomics to enrich and identify the glutathionylated peptides in Escherichia coli and Drosophila lysates, in which 937 and 1,930 potential glutathionylated peptides were identified, respectively. Bioinformatics analysis showed that the cysteine residue next to negatively charged amino acid residues has a higher frequency of glutathionylation. Importantly, we found that most proteins associated with metabolic pathways were glutathionylated and that the glutathionylation sites of metabolic enzymes were highly conserved among different species. Our results indicate that the glutathione analog is a useful tool to characterize protein glutathionylation, and glutathionylation of metabolic enzymes, which play important roles in regulating cellular metabolism, is conserved.
Collapse
|
67
|
Wilson C, González-Billault C. Regulation of cytoskeletal dynamics by redox signaling and oxidative stress: implications for neuronal development and trafficking. Front Cell Neurosci 2015; 9:381. [PMID: 26483635 PMCID: PMC4588006 DOI: 10.3389/fncel.2015.00381] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/11/2015] [Indexed: 01/10/2023] Open
Abstract
A proper balance between chemical reduction and oxidation (known as redox balance) is essential for normal cellular physiology. Deregulation in the production of oxidative species leads to DNA damage, lipid peroxidation and aberrant post-translational modification of proteins, which in most cases induces injury, cell death and disease. However, physiological concentrations of oxidative species are necessary to support important cell functions, such as chemotaxis, hormone synthesis, immune response, cytoskeletal remodeling, Ca2+ homeostasis and others. Recent evidence suggests that redox balance regulates actin and microtubule dynamics in both physiological and pathological contexts. Microtubules and actin microfilaments contain certain amino acid residues that are susceptible to oxidation, which reduces the ability of microtubules to polymerize and causes severing of actin microfilaments in neuronal and non-neuronal cells. In contrast, inhibited production of reactive oxygen species (ROS; e.g., due to NOXs) leads to aberrant actin polymerization, decreases neurite outgrowth and affects the normal development and polarization of neurons. In this review, we summarize emerging evidence suggesting that both general and specific enzymatic sources of redox species exert diverse effects on cytoskeletal dynamics. Considering the intimate relationship between cytoskeletal dynamics and trafficking, we also discuss the potential effects of redox balance on intracellular transport via regulation of the components of the microtubule and actin cytoskeleton as well as cytoskeleton-associated proteins, which may directly impact localization of proteins and vesicles across the soma, dendrites and axon of neurons.
Collapse
Affiliation(s)
- Carlos Wilson
- Department of Biology, Faculty of Sciences, Universidad de Chile Santiago, Chile
| | | |
Collapse
|
68
|
Kotz S, Kullmann M, Crone B, Kalayda GV, Jaehde U, Metzger S. Combination of two-dimensional gel electrophoresis and a fluorescent carboxyfluorescein-diacetate-labeled cisplatin analogue allows the identification of intracellular cisplatin-protein adducts. Electrophoresis 2015; 36:2811-2819. [DOI: 10.1002/elps.201500188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 06/27/2015] [Accepted: 07/13/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Sandra Kotz
- Biocenter MS Platform, Department of Biology; University of Cologne; Cologne Germany
| | - Maximilian Kullmann
- Department of Clinical Pharmacy, Institute of Pharmacy; University of Bonn; Bonn Germany
| | - Barbara Crone
- Institute of Inorganic and Analytical Chemistry; University of Münster; Münster Germany
| | - Ganna V. Kalayda
- Department of Clinical Pharmacy, Institute of Pharmacy; University of Bonn; Bonn Germany
| | - Ulrich Jaehde
- Department of Clinical Pharmacy, Institute of Pharmacy; University of Bonn; Bonn Germany
| | - Sabine Metzger
- Biocenter MS Platform, Department of Biology; University of Cologne; Cologne Germany
- IUF-Leibniz Research Institute for Environmental Medicine; IUF; Düsseldorf Germany
| |
Collapse
|
69
|
Khaitlina SY. Tropomyosin as a Regulator of Actin Dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 318:255-91. [PMID: 26315888 DOI: 10.1016/bs.ircmb.2015.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Tropomyosin is a major regulatory protein of contractile systems and cytoskeleton, an actin-binding protein that positions laterally along actin filaments and modulates actin-myosin interaction. About 40 tropomyosin isoforms have been found in a variety of cytoskeleton systems, not necessarily connected with actin-myosin interaction and contraction. Involvement of specific tropomyosin isoforms in the regulation of key cell processes was shown, and specific features of tropomyosin genes and protein structure have been investigated with molecular biology and genetics approaches. However, the mechanisms underlying the effects of tropomyosin on cytoskeleton dynamics are still unclear. As tropomyosin is primarily an F-actin-binding protein, it is important to understand how it interacts both with actin and actin-binding proteins functioning in muscles and cytoskeleton to regulate actin dynamics. This review focuses on biochemical data on the effects of tropomyosin on actin assembly and dynamics, as well as on the modulation of these effects by actin-binding proteins. The data indicate that tropomyosin can efficiently regulate actin dynamics via allosteric conformational changes within actin filaments.
Collapse
Affiliation(s)
- Sofia Yu Khaitlina
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia.
| |
Collapse
|
70
|
Breitkreuz M, Hamdani N. A change of heart: oxidative stress in governing muscle function? Biophys Rev 2015; 7:321-341. [PMID: 28510229 DOI: 10.1007/s12551-015-0175-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023] Open
Abstract
Redox/cysteine modification of proteins that regulate calcium cycling can affect contraction in striated muscles. Understanding the nature of these modifications would present the possibility of enhancing cardiac function through reversible cysteine modification of proteins, with potential therapeutic value in heart failure with diastolic dysfunction. Both heart failure and muscular dystrophy are characterized by abnormal redox balance and nitrosative stress. Recent evidence supports the synergistic role of oxidative stress and inflammation in the progression of heart failure with preserved ejection fraction, in concert with endothelial dysfunction and impaired nitric oxide-cyclic guanosine monophosphate-protein kinase G signalling via modification of the giant protein titin. Although antioxidant therapeutics in heart failure with diastolic dysfunction have no marked beneficial effects on the outcome of patients, it, however, remains critical to the understanding of the complex interactions of oxidative/nitrosative stress with pro-inflammatory mechanisms, metabolic dysfunction, and the redox modification of proteins characteristic of heart failure. These may highlight novel approaches to therapeutic strategies for heart failure with diastolic dysfunction. In this review, we provide an overview of oxidative stress and its effects on pathophysiological pathways. We describe the molecular mechanisms driving oxidative modification of proteins and subsequent effects on contractile function, and, finally, we discuss potential therapeutic opportunities for heart failure with diastolic dysfunction.
Collapse
Affiliation(s)
- Martin Breitkreuz
- Department of Cardiovascular Physiology, Ruhr University Bochum, MA 3/56, 44780, Bochum, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, MA 3/56, 44780, Bochum, Germany.
| |
Collapse
|
71
|
Fang Z, Dos Santos PC. Protective role of bacillithiol in superoxide stress and Fe-S metabolism in Bacillus subtilis. Microbiologyopen 2015; 4:616-31. [PMID: 25988368 PMCID: PMC4554457 DOI: 10.1002/mbo3.267] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 01/22/2023] Open
Abstract
Glutathione (GSH) serves as the prime thiol in most organisms as its depletion increases antibiotic and metal toxicity, impairs oxidative stress responses, and affects Fe and Fe–S cluster metabolism. Many gram-positive bacteria lack GSH, but instead produce other structurally unrelated yet functionally equivalent thiols. Among those, bacillithiol (BSH) has been recently identified in several low G+C gram-positive bacteria. In this work, we have explored the link between BSH and Fe–S metabolism in Bacillus subtilis. We have identified that B. subtilis lacking BSH is more sensitive to oxidative stress (paraquat), and metal toxicity (Cu(I) and Cd(II)), but not H2O2. Furthermore, a slow growth phenotype of BSH null strain in minimal medium was observed, which could be recovered upon the addition of selected amino acids (Leu/Ile and Glu/Gln), supplementation of iron, or chemical complementation with BSH disulfide (BSSB) to the growth medium. Interestingly, Fe–S cluster containing isopropylmalate isomerase (LeuCD) and glutamate synthase (GOGAT) showed decreased activities in BSH null strain. Deficiency of BSH also resulted in decreased levels of intracellular Fe accompanied by increased levels of manganese and altered expression levels of Fe–S cluster biosynthetic SUF components. Together, this study is the first to establish a link between BSH and Fe–S metabolism in B. subtilis.
Collapse
Affiliation(s)
- Zhong Fang
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, 27016
| | - Patricia C Dos Santos
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, 27016
| |
Collapse
|
72
|
Sulfhydryl-mediated redox signaling in inflammation: role in neurodegenerative diseases. Arch Toxicol 2015; 89:1439-67. [DOI: 10.1007/s00204-015-1496-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/25/2015] [Indexed: 01/05/2023]
|
73
|
Siu AW, Shan SW, Li KK, Lam HY, Fung MY, Li KK, To CH, Do CW. Glutathione attenuates nitric oxide-induced retinal lipid and protein changes. Ophthalmic Physiol Opt 2015; 35:135-46. [DOI: 10.1111/opo.12198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 12/30/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Andrew W. Siu
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Sze Wan Shan
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - King Kit Li
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Hiu Yan Lam
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Man Yee Fung
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Ka Ki Li
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Chi Ho To
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou China
| | - Chi Wai Do
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| |
Collapse
|
74
|
Rothmeier AS, Marchese P, Petrich BG, Furlan-Freguia C, Ginsberg MH, Ruggeri ZM, Ruf W. Caspase-1-mediated pathway promotes generation of thromboinflammatory microparticles. J Clin Invest 2015; 125:1471-84. [PMID: 25705884 DOI: 10.1172/jci79329] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/09/2015] [Indexed: 12/30/2022] Open
Abstract
Extracellular ATP is a signal of tissue damage and induces macrophage responses that amplify inflammation and coagulation. Here we demonstrate that ATP signaling through macrophage P2X7 receptors uncouples the thioredoxin (TRX)/TRX reductase (TRXR) system and activates the inflammasome through endosome-generated ROS. TRXR and inflammasome activity promoted filopodia formation, cellular release of reduced TRX, and generation of extracellular thiol pathway-dependent, procoagulant microparticles (MPs). Additionally, inflammasome-induced activation of an intracellular caspase-1/calpain cysteine protease cascade degraded filamin, thereby severing bonds between the cytoskeleton and tissue factor (TF), the cell surface receptor responsible for coagulation activation. This cascade enabled TF trafficking from rafts to filopodia and ultimately onto phosphatidylserine-positive, highly procoagulant MPs. Furthermore, caspase-1 specifically facilitated cell surface actin exposure, which was required for the final release of highly procoagulant MPs from filopodia. Together, the results of this study delineate a thromboinflammatory pathway and suggest that components of this pathway have potential as pharmacological targets to simultaneously attenuate inflammation and innate immune cell-induced thrombosis.
Collapse
|
75
|
Beckendorf L, Linke WA. Emerging importance of oxidative stress in regulating striated muscle elasticity. J Muscle Res Cell Motil 2014; 36:25-36. [PMID: 25373878 PMCID: PMC4352196 DOI: 10.1007/s10974-014-9392-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/03/2014] [Indexed: 12/11/2022]
Abstract
The contractile function of striated muscle cells is altered by oxidative/nitrosative stress, which can be observed under physiological conditions but also in diseases like heart failure or muscular dystrophy. Oxidative stress causes oxidative modifications of myofilament proteins and can impair myocyte contractility. Recent evidence also suggests an important effect of oxidative stress on muscle elasticity and passive stiffness via modifications of the giant protein titin. In this review we provide a short overview of known oxidative modifications in thin and thick filament proteins and then discuss in more detail those oxidative stress-related modifications altering titin stiffness directly or indirectly. Direct modifications of titin include reversible disulfide bonding within the cardiac-specific N2-Bus domain, which increases titin stiffness, and reversible S-glutathionylation of cryptic cysteines in immunoglobulin-like domains, which only takes place after the domains have unfolded and which reduces titin stiffness in cardiac and skeletal muscle. Indirect effects of oxidative stress on titin can occur via reversible modifications of protein kinase signalling pathways (especially the NO-cGMP-PKG axis), which alter the phosphorylation level of certain disordered titin domains and thereby modulate titin stiffness. Oxidative stress also activates proteases such as matrix-metalloproteinase-2 and (indirectly via increasing the intracellular calcium level) calpain-1, both of which cleave titin to irreversibly reduce titin-based stiffness. Although some of these mechanisms require confirmation in the in vivo setting, there is evidence that oxidative stress-related modifications of titin are relevant in the context of biomarker design and represent potential targets for therapeutic intervention in some forms of muscle and heart disease.
Collapse
Affiliation(s)
- Lisa Beckendorf
- Department of Cardiovascular Physiology, Institute of Physiology, Ruhr University Bochum, MA 3/56, 44780, Bochum, Germany
| | | |
Collapse
|
76
|
Luo W, Lin B, Wang Y, Zhong J, O'Meally R, Cole RN, Pandey A, Levchenko A, Semenza GL. PHD3-mediated prolyl hydroxylation of nonmuscle actin impairs polymerization and cell motility. Mol Biol Cell 2014; 25:2788-96. [PMID: 25079693 PMCID: PMC4161513 DOI: 10.1091/mbc.e14-02-0775] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 07/10/2014] [Accepted: 07/10/2014] [Indexed: 01/03/2023] Open
Abstract
Actin filaments play an essential role in cell movement, and many posttranslational modifications regulate actin filament assembly. Here we report that prolyl hydroxylase 3 (PHD3) interacts with nonmuscle actin in human cells and catalyzes hydroxylation of actin at proline residues 307 and 322. Blocking PHD3 expression or catalytic activity by short hairpin RNA knockdown or pharmacological inhibition, respectively, decreased actin prolyl hydroxylation. PHD3 knockdown increased filamentous F-actin assembly, which was reversed by PHD3 overexpression. PHD3 knockdown increased cell velocity and migration distance. Inhibition of PHD3 prolyl hydroxylase activity by dimethyloxalylglycine also increased actin polymerization and cell migration. These data reveal a novel role for PHD3 as a negative regulator of cell motility through posttranslational modification of nonmuscle actins.
Collapse
Affiliation(s)
- Weibo Luo
- Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Benjamin Lin
- Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Yingfei Wang
- Neuroregeneration Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jun Zhong
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Robert O'Meally
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Robert N Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Akhilesh Pandey
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Andre Levchenko
- Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Gregg L Semenza
- Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
77
|
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]
|
78
|
Abstract
SIGNIFICANCE Adhesion and migration induced by cytokines or growth factors are well-organized processes in cellular motility. Reactive oxygen species (ROS) are specifically produced by the Nox family of NADPH oxidases. RECENT ADVANCES The signal transduction of migration and adhesion depends on ROS produced by Nox enzymes and factors that initiate migration and adhesion and stimulate cellular ROS formation. CRITICAL ISSUES The identification of molecular targets of ROS formation in the signal transduction of adhesion and migration is still in its beginnings, but a site and isoform-specific contribution of Nox enzymes to this process becomes apparent. Nox-derived ROS, therefore, act as second messengers that are specifically modifying signaling proteins involved in adhesion and migration. FUTURE DIRECTIONS Individual protein targets of Nox-mediated redox signaling in different cell types and tissues will be identified. Isoform-specific Nox inhibitors will be developed to modulate the ROS-dependent component of migration and adhesion. These compounds might be suited to elicit differential effects between pathophysiologic and physiologic adhesion and migration.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität , Frankfurt am Main, Germany
| |
Collapse
|
79
|
Oxidative stress in muscular dystrophy: from generic evidence to specific sources and targets. J Muscle Res Cell Motil 2014; 35:23-36. [PMID: 24619215 DOI: 10.1007/s10974-014-9380-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 02/19/2014] [Indexed: 01/06/2023]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases that share a common end-point represented by muscular wasting. MDs are caused by mutations in a variety of genes encoding for different molecules, including extracellular matrix, transmembrane and membrane-associated proteins, cytoplasmic enzymes and nuclear proteins. However, it is still to be elucidated how genetic mutations can affect the molecular mechanisms underlying the contractile impairment occurring in these complex pathologies. The intracellular accumulation of reactive oxygen species (ROS) is widely accepted to play a key role in contractile derangements occurring in the different forms of MDs. However, scarce information is available concerning both the most relevant sources of ROS and their major molecular targets. This review focuses on (i) the sources of ROS, with a special emphasis on monoamine oxidase, a mitochondrial enzyme, and (ii) the targets of ROS, highlighting the relevance of the oxidative modification of myofilament proteins.
Collapse
|
80
|
Sobierajska K, Skurzynski S, Stasiak M, Kryczka J, Cierniewski CS, Swiatkowska M. Protein disulfide isomerase directly interacts with β-actin Cys374 and regulates cytoskeleton reorganization. J Biol Chem 2014; 289:5758-73. [PMID: 24415753 DOI: 10.1074/jbc.m113.479477] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent studies support the role of cysteine oxidation in actin cytoskeleton reorganization during cell adhesion. The aim of this study was to explain whether protein disulfide isomerase (PDI) is responsible for the thiol-disulfide rearrangement in the β-actin molecule of adhering cells. First, we showed that PDI forms a disulfide-bonded complex with β-actin with a molecular mass of 110 kDa. Specific interaction of both proteins was demonstrated by a solid phase binding assay, surface plasmon resonance analysis, and immunoprecipitation experiments. Second, using confocal microscopy, we found that both proteins colocalized when spreading MEG-01 cells on fibronectin. Colocalization of PDI and β-actin could be abolished by the membrane-permeable sulfhydryl blocker, N-ethylmaleimide, by the RGD peptide, and by anti-αIIbβ3 antibodies. Consequently, down-regulation of PDI expression by antisense oligonucleotides impaired the spreading of cells and initiated reorganization of the cytoskeleton. Third, because of transfection experiments followed by immunoprecipitation and confocal analysis, we provided evidence that PDI binds to the β-actin Cys(374) thiol. Formation of the β-actin-PDI complex was mediated by integrin-dependent signaling in response to the adhesion of cells to the extracellular matrix. Our data suggest that PDI is released from subcellular compartments to the cytosol and translocated toward the periphery of the cell, where it forms a disulfide bond with β-actin when MEG-01 cells adhere via the αIIbβ3 integrin to fibronectin. Thus, PDI appears to regulate cytoskeletal reorganization by the thiol-disulfide exchange in β-actin via a redox-dependent mechanism.
Collapse
Affiliation(s)
- Katarzyna Sobierajska
- From the Department of Molecular and Medical Biophysics, Medical University of Lodz, 92-215 Lodz, Poland and
| | | | | | | | | | | |
Collapse
|
81
|
Patel BG, Wilder T, Solaro RJ. Novel control of cardiac myofilament response to calcium by S-glutathionylation at specific sites of myosin binding protein C. Front Physiol 2013; 4:336. [PMID: 24312057 PMCID: PMC3834529 DOI: 10.3389/fphys.2013.00336] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/01/2013] [Indexed: 12/31/2022] Open
Abstract
Our previous studies demonstrated a relation between glutathionylation of cardiac myosin binding protein C (cMyBP-C) and diastolic dysfunction in a hypertensive mouse model stressed by treatment with salt, deoxycorticosterone acetate, and unilateral nephrectomy. Although these results strongly indicated an important role for S-glutathionylation of myosin binding protein C as a modifier of myofilament function, indirect effects of other post-translational modifications may have occurred. Moreover, we did not determine the sites of thiol modification by glutathionylation. To address these issues, we developed an in vitro method to mimic the in situ S-glutathionylation of myofilament proteins and determined direct functional effects and sites of oxidative modification employing Western blotting and mass spectrometry. We induced glutathionylation in vitro by treatment of isolated myofibrils and detergent extracted fiber bundles (skinned fibers) with oxidized glutathione (GSSG). Immuno-blotting results revealed increased glutathionylation with GSSG treatment of a protein band around 140 kDa. Using tandem mass spectrometry, we identified the 140 kDa band as cMyBP-C and determined the sites of glutathionylation to be at cysteines 655, 479, and 627. Determination of the relation between Ca2+-activation of myofibrillar acto-myosin ATPase rate demonstrated an increased Ca2+-sensitivity induced by the S-glutathionylation. Force generating skinned fiber bundles also showed an increase in Ca-sensitivity when treated with oxidized glutathione, which was reversed with the reducing agent, dithiothreitol (DTT). Our data demonstrate that a specific and direct effect of S-glutathionylation of myosin binding protein C is a significant increase in myofilament Ca2+-sensitivity. Our data also provide new insights into the functional significance of oxidative modification of myosin binding protein C and the potential role of domains not previously considered to be functionally significant as controllers of myofilament Ca2+-responsiveness and dynamics.
Collapse
Affiliation(s)
- Bindiya G Patel
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago Chicago, IL USA
| | | | | |
Collapse
|
82
|
Pastore A, Piemonte F. Protein glutathionylation in cardiovascular diseases. Int J Mol Sci 2013; 14:20845-76. [PMID: 24141185 PMCID: PMC3821647 DOI: 10.3390/ijms141020845] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/02/2013] [Accepted: 10/08/2013] [Indexed: 02/07/2023] Open
Abstract
The perturbation of thiol-disulfide homeostasis is an important consequence of many diseases, with redox signals implicated in several physio-pathological processes. A prevalent form of cysteine modification is the reversible formation of protein mixed disulfides with glutathione (S-glutathionylation). The abundance of glutathione in cells and the ready conversion of sulfenic acids to S-glutathione mixed disulfides supports the reversible protein S-glutathionylation as a common feature of redox signal transduction, able to regulate the activities of several redox sensitive proteins. In particular, protein S-glutathionylation is emerging as a critical signaling mechanism in cardiovascular diseases, because it regulates numerous physiological processes involved in cardiovascular homeostasis, including myocyte contraction, oxidative phosphorylation, protein synthesis, vasodilation, glycolytic metabolism and response to insulin. Thus, perturbations in protein glutathionylation status may contribute to the etiology of many cardiovascular diseases, such as myocardial infarction, cardiac hypertrophy and atherosclerosis. Various reports show the importance of oxidative cysteine modifications in modulating cardiovascular function. In this review, we illustrate tools and strategies to monitor protein S-glutathionylation and describe the proteins so far identified as glutathionylated in myocardial contraction, hypertrophy and inflammation.
Collapse
Affiliation(s)
- Anna Pastore
- Laboratory of Biochemistry, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; E-Mail:
| | - Fiorella Piemonte
- Unit of Neuromuscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
83
|
Terrill JR, Boyatzis A, Grounds MD, Arthur PG. Treatment with the cysteine precursor l-2-oxothiazolidine-4-carboxylate (OTC) implicates taurine deficiency in severity of dystropathology in mdx mice. Int J Biochem Cell Biol 2013; 45:2097-108. [PMID: 23892094 DOI: 10.1016/j.biocel.2013.07.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/28/2013] [Accepted: 07/09/2013] [Indexed: 01/29/2023]
Abstract
Oxidative stress has been implicated in the pathology of the lethal skeletal muscle disease Duchenne muscular dystrophy (DMD), and various antioxidants have been investigated as a potential therapy. Recently, treatment of the mdx mouse model for DMD with the antioxidant and cysteine and glutathione (GSH) precursor n-acetylcysteine (NAC) was shown to decrease protein thiol oxidation and improve muscle pathology and ex vivo muscle strength. This study further investigates the mechanism for the benefits of NAC on dystrophic muscle by administering l-2-oxothiazolidine-4-carboxylate (OTC) which also upregulates intracellular cysteine and GSH, but does not directly function as an antioxidant. We observed that OTC, like NAC, decreases protein thiol oxidation, decreases pathology and increases strength, suggesting that the both NAC and OTC function via increasing cysteine and GSH content of dystrophic muscle. We demonstrate that mdx muscle is not deficient in either cysteine or GSH and that these are not increased by OTC treatment. However, we show that dystrophic muscle of 12 week old mdx mice is deficient in taurine, a by-product of disposal of excess cysteine, a deficiency that is ameliorated by OTC treatment. These data suggest that in dystrophic muscles, apart from the strong association of increased oxidative stress and protein thiol oxidation with dystropathology, another major issue is an insufficiency in taurine that can be corrected by increasing the availability of cysteine. This study provides new insight into the molecular mechanism underlying the benefits of NAC in muscular dystrophy and supports the use of OTC as an alternative drug for potential clinical applications to DMD.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia, Australia.
| | | | | | | |
Collapse
|
84
|
Ullevig S, Kim HS, Asmis R. S-glutathionylation in monocyte and macrophage (dys)function. Int J Mol Sci 2013; 14:15212-32. [PMID: 23887649 PMCID: PMC3759857 DOI: 10.3390/ijms140815212] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 06/15/2013] [Accepted: 06/18/2013] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease involving the accumulation of monocytes and macrophages in the vascular wall. Monocytes and macrophages play a central role in the initiation and progression of atherosclerotic lesion development. Oxidative stress, which occurs when reactive oxygen species (ROS) overwhelm cellular antioxidant systems, contributes to the pathophysiology of many chronic inflammatory diseases, including atherosclerosis. Major targets of ROS are reactive thiols on cysteine residues in proteins, which when oxidized can alter cellular processes, including signaling pathways, metabolic pathways, transcription, and translation. Protein-S-glutathionylation is the process of mixed disulfide formation between glutathione (GSH) and protein thiols. Until recently, protein-S-glutathionylation was associated with increased cellular oxidative stress, but S-glutathionylation of key protein targets has now emerged as a physiologically important redox signaling mechanism, which when dysregulated contributes to a variety of disease processes. In this review, we will explore the role of thiol oxidative stress and protein-S-glutathionylation in monocyte and macrophage dysfunction as a mechanistic link between oxidative stress associated with metabolic disorders and chronic inflammatory diseases, including atherosclerosis.
Collapse
Affiliation(s)
- Sarah Ullevig
- Department of Biochemistry, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
| | - Hong Seok Kim
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
| | - Reto Asmis
- Department of Biochemistry, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-210-567-3411; Fax: +1-210-567-3719
| |
Collapse
|
85
|
Choong G, Liu Y, Xiao W, Templeton DM. Cadmium-induced glutathionylation of actin occurs through a ROS-independent mechanism: implications for cytoskeletal integrity. Toxicol Appl Pharmacol 2013; 272:423-30. [PMID: 23872096 DOI: 10.1016/j.taap.2013.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/27/2013] [Accepted: 07/05/2013] [Indexed: 12/27/2022]
Abstract
Cadmium disrupts the actin cytoskeleton in rat mesangial cells, and we have previously shown that this involves a complex interplay involving activation of kinase signaling, protein translocation, and disruption of focal adhesions. Here we investigate the role that glutathionylation of actin plays in Cd(2+)-associated cytoskeletal reorganization. Low concentrations of Cd(2+) (0.5-2 μM) caused an increase in actin glutathionylation by 6h, whereas at higher concentrations glutathionylation remained at basal levels. Although oxidation with diamide increased glutathionylation, reactive oxygen species (ROS) were not involved in the Cd(2+)-dependent effect, as only Cd(2+) concentrations above 2 μM were sufficient to increase ROS. However, low [Cd(2+)] increased total glutathione levels without affecting the ratio of reduced/oxidized glutathione, and inhibition of glutathione synthesis suppressed actin glutathionylation. Cadmium increased the activity of the enzyme glutaredoxin, which influences the equilibrium between glutathionylated and deglutathionylated proteins and thus may influence levels of glutathionylated actin. Together these observations show that cadmium-dependent effects on actin glutathionylation are affected by glutathione metabolism and not by direct effects of ROS on thiol chemistry. In vitro polymerization assays with glutathionylated actin show a decreased rate of polymerization. In contrast, immunofluorescence of cytoskeletal structure in intact cells suggests that increases in actin glutathionylation accompanying increased glutathione levels occurring under low Cd(2+) exposure are protective in vivo, with cytoskeletal disruption ensuing only when higher Cd(2+) concentrations increase ROS levels and prevent an increase in actin-glutathione conjugates.
Collapse
Affiliation(s)
- Grace Choong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada
| | | | | | | |
Collapse
|
86
|
Terrill JR, Radley-Crabb HG, Iwasaki T, Lemckert FA, Arthur PG, Grounds MD. Oxidative stress and pathology in muscular dystrophies: focus on protein thiol oxidation and dysferlinopathies. FEBS J 2013; 280:4149-64. [PMID: 23332128 DOI: 10.1111/febs.12142] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 12/23/2022]
Abstract
The muscular dystrophies comprise more than 30 clinical disorders that are characterized by progressive skeletal muscle wasting and degeneration. Although the genetic basis for many of these disorders has been identified, the exact mechanism for pathogenesis generally remains unknown. It is considered that disturbed levels of reactive oxygen species (ROS) contribute to the pathology of many muscular dystrophies. Reactive oxygen species and oxidative stress may cause cellular damage by directly and irreversibly damaging macromolecules such as proteins, membrane lipids and DNA; another major cellular consequence of reactive oxygen species is the reversible modification of protein thiol side chains that may affect many aspects of molecular function. Irreversible oxidative damage of protein and lipids has been widely studied in Duchenne muscular dystrophy, and we have recently identified increased protein thiol oxidation in dystrophic muscles of the mdx mouse model for Duchenne muscular dystrophy. This review evaluates the role of elevated oxidative stress in Duchenne muscular dystrophy and other forms of muscular dystrophies, and presents new data that show significantly increased protein thiol oxidation and high levels of lipofuscin (a measure of cumulative oxidative damage) in dysferlin-deficient muscles of A/J mice at various ages. The significance of this elevated oxidative stress and high levels of reversible thiol oxidation, but minimal myofibre necrosis, is discussed in the context of the disease mechanism for dysferlinopathies, and compared with the situation for dystrophin-deficient mdx mice.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
87
|
Protein sulfhydryl group oxidation and mixed-disulfide modifications in stable and unstable human carotid plaques. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:403973. [PMID: 23431411 PMCID: PMC3575616 DOI: 10.1155/2013/403973] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 12/28/2012] [Indexed: 11/17/2022]
Abstract
Objectives. Oxidative stress has been implicated in the outcome of atherosclerotic plaques. However, at present, no data are available neither on the degree of plaque protein sulfhydryl groups oxidation nor on its relationship with plaque vulnerability. We investigated the entity of protein-SH oxidative modifications, focusing on low molecular weight thiols adduction, in human carotid plaque extracts in relation to plaque stability/instability. Methods. Plaque stability/instability was histologically assessed. The extent of protein-SH oxidative modifications was established by a differential proteomic approach on fluorescein-5-maleimide-labeled plaque extracts and corresponding plasma samples from 48 endarterectomized patients. The analysis on protein thiolation was performed by capillary zone electrophoresis. Results. We observed a higher protein-SH oxidation of both plasma-derived and topically expressed proteins in unstable plaques, partly due to higher levels of S-thiolation. Conversely, in plasma, none of the investigated parameters discriminated among patients with stable and unstable plaques. Conclusions. Our results suggest the presence of a more pronounced oxidative environment in unstable plaques. Identifying specific oxidative modifications and understanding their effects on protein function could provide further insight into the relevance of oxidative stress in atherosclerosis.
Collapse
|
88
|
Chung HS, Wang SB, Venkatraman V, Murray CI, Van Eyk JE. Cysteine oxidative posttranslational modifications: emerging regulation in the cardiovascular system. Circ Res 2013; 112:382-92. [PMID: 23329793 PMCID: PMC4340704 DOI: 10.1161/circresaha.112.268680] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Accepted: 10/05/2012] [Indexed: 12/22/2022]
Abstract
In the cardiovascular system, changes in oxidative balance can affect many aspects of cellular physiology through redox-signaling. Depending on the magnitude, fluctuations in the cell's production of reactive oxygen and nitrogen species can regulate normal metabolic processes, activate protective mechanisms, or be cytotoxic. Reactive oxygen and nitrogen species can have many effects including the posttranslational modification of proteins at critical cysteine thiols. A subset can act as redox-switches, which elicit functional effects in response to changes in oxidative state. Although the general concepts of redox-signaling have been established, the identity and function of many regulatory switches remains unclear. Characterizing the effects of individual modifications is the key to understand how the cell interprets oxidative signals under physiological and pathological conditions. Here, we review the various cysteine oxidative posttranslational modifications and their ability to function as redox-switches that regulate the cell's response to oxidative stimuli. In addition, we discuss how these modifications have the potential to influence other posttranslational modifications' signaling pathways though cross-talk. Finally, we review the increasing number of tools being developed to identify and quantify the various cysteine oxidative posttranslational modifications and how this will advance our understanding of redox-regulation.
Collapse
Affiliation(s)
- Heaseung S Chung
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
89
|
Rodriguez-Rocha H, Garcia Garcia A, Zavala-Flores L, Li S, Madayiputhiya N, Franco R. Glutaredoxin 1 protects dopaminergic cells by increased protein glutathionylation in experimental Parkinson's disease. Antioxid Redox Signal 2012; 17:1676-93. [PMID: 22816731 PMCID: PMC3474191 DOI: 10.1089/ars.2011.4474] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Chronic exposure to environmental toxicants, such as paraquat, has been suggested as a risk factor for Parkinson's disease (PD). Although dopaminergic cell death in PD is associated with oxidative damage, the molecular mechanisms involved remain elusive. Glutaredoxins (GRXs) utilize the reducing power of glutathione to modulate redox-dependent signaling pathways by protein glutathionylation. We aimed to determine the role of GRX1 and protein glutathionylation in dopaminergic cell death. RESULTS In dopaminergic cells, toxicity induced by paraquat or 6-hydroxydopamine (6-OHDA) was inhibited by GRX1 overexpression, while its knock-down sensitized cells to paraquat-induced cell death. Dopaminergic cell death was paralleled by protein deglutathionylation, and this was reversed by GRX1. Mass spectrometry analysis of immunoprecipitated glutathionylated proteins identified the actin binding flightless-1 homolog protein (FLI-I) and the RalBP1-associated Eps domain-containing protein 2 (REPS2/POB1) as targets of glutathionylation in dopaminergic cells. Paraquat induced the degradation of FLI-I and REPS2 proteins, which corresponded with the activation of caspase 3 and cell death progression. GRX1 overexpression reduced both the degradation and deglutathionylation of FLI-I and REPS2, while stable overexpression of REPS2 reduced paraquat toxicity. A decrease in glutathionylated proteins and REPS2 levels was also observed in the substantia nigra of mice treated with paraquat. INNOVATION We have identified novel protein targets of glutathionylation in dopaminergic cells and demonstrated the protective role of GRX1-mediated protein glutathionylation against paraquat-induced toxicity. CONCLUSIONS These results demonstrate a protective role for GRX1 and increased protein glutathionylation in dopaminergic cell death induced by paraquat, and identify a novel protective role for REPS2.
Collapse
|
90
|
Identification of proteins containing redox-sensitive thiols after PRDX1, PRDX3 and GCLC silencing and/or glucose oxidase treatment in Hepa 1–6 cells. J Proteomics 2012; 77:262-79. [DOI: 10.1016/j.jprot.2012.08.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/07/2012] [Accepted: 08/22/2012] [Indexed: 12/20/2022]
|
91
|
Bachi A, Dalle-Donne I, Scaloni A. Redox Proteomics: Chemical Principles, Methodological Approaches and Biological/Biomedical Promises. Chem Rev 2012. [DOI: 10.1021/cr300073p] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Angela Bachi
- Biological Mass Spectrometry Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy
| |
Collapse
|
92
|
Sakai J, Li J, Subramanian KK, Mondal S, Bajrami B, Hattori H, Jia Y, Dickinson BC, Zhong J, Ye K, Chang CJ, Ho YS, Zhou J, Luo HR. Reactive oxygen species-induced actin glutathionylation controls actin dynamics in neutrophils. Immunity 2012; 37:1037-49. [PMID: 23159440 DOI: 10.1016/j.immuni.2012.08.017] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 08/21/2012] [Indexed: 12/29/2022]
Abstract
The regulation of actin dynamics is pivotal for cellular processes such as cell adhesion, migration, and phagocytosis and thus is crucial for neutrophils to fulfill their roles in innate immunity. Many factors have been implicated in signal-induced actin polymerization, but the essential nature of the potential negative modulators are still poorly understood. Here we report that NADPH oxidase-dependent physiologically generated reactive oxygen species (ROS) negatively regulate actin polymerization in stimulated neutrophils via driving reversible actin glutathionylation. Disruption of glutaredoxin 1 (Grx1), an enzyme that catalyzes actin deglutathionylation, increased actin glutathionylation, attenuated actin polymerization, and consequently impaired neutrophil polarization, chemotaxis, adhesion, and phagocytosis. Consistently, Grx1-deficient murine neutrophils showed impaired in vivo recruitment to sites of inflammation and reduced bactericidal capability. Together, these results present a physiological role for glutaredoxin and ROS- induced reversible actin glutathionylation in regulation of actin dynamics in neutrophils.
Collapse
Affiliation(s)
- Jiro Sakai
- Department of Pathology, Harvard Medical School and Department of Lab Medicine, Children's Hospital Boston and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Eligini S, Songia P, Cavalca V, Crisci M, Tremoli E, Colli S. Cytoskeletal architecture regulates cyclooxygenase-2 in human endothelial cells: autocrine modulation by prostacyclin. J Cell Physiol 2012; 227:3847-56. [PMID: 22495438 DOI: 10.1002/jcp.24097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelium is a highly dynamic tissue that controls vascular homeostasis. This requires constant rearrangements of the shape or function of endothelial cells that cannot set aside the role of the cytoskeleton. The aim of this study was to determine the mechanisms by means of which cytoskeletal alterations induce cyclooxygenase-2 (Cox-2) expression in human endothelial cells using compounds that interfere with microtubule or actin architecture. Microtubule disruption by nocodazole markedly increased Cox-2 expression and activity, and provoked paracellular gap formation, a cardinal feature of endothelial barrier dysfunction. The Cox-2 metabolite prostacyclin down-regulated Cox-2 through an autocrine receptor-mediated mechanism, and partially prevented the disassembly of endothelial monolayers. There was also an interaction between microtubules and actin filaments in nocodazole-induced Cox-2 expression. Nocodazole provoked the dissolution of the F-actin cortical ring and stress fiber formation, increased actin glutathionylation, and concomitantly lowered intracellular levels of reduced glutathione. The restoration of glutathione levels by N-acetylcysteine opposed Cox-2 expression and preserved the integrity of endothelial monolayers. Among the signaling pathways connecting microtubule disruption with Cox-2 up-regulation, crucial roles are played by Src family kinase activation, serine/threonine phosphatase 2A inhibition, and the phosphorylation of mitogen activated protein kinase p38. Our findings provide a mechanistic insight into the observation that Cox-2 is induced in endothelial cells under cytoskeleton-perturbing conditions such as those occurring in the presence of atherogenic/inflammatory stimuli and oxidative stress. In this scenario, Cox-2 up-regulation by endothelia exposed to noxious conditions can be considered protective of the vasodilatory and anti-thrombotic properties of the vessel wall.
Collapse
|
94
|
Gödecke A, Schrader J, Reinartz M. Nitric oxide-mediated protein modification in cardiovascular physiology and pathology. Proteomics Clin Appl 2012; 2:811-22. [PMID: 21136881 DOI: 10.1002/prca.200780079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO) is a key regulator of cardiovascular functions including the control of vascular tone, anti-inflammatory properties of the endothelium, cardiac contractility, and thrombocyte activation and aggregation. Numerous experimental data support the view that NO not only acts via cyclic guanosine monophosphate (cGMP)-dependent mechanisms but also modulates protein function by nitrosation, nitrosylation, glutathiolation, and nitration, respectively. To understand how NO regulates all of these diverse biological processes on the molecular level a comprehensive assessment of NO-mediated cGMP-dependent and independent targets is required. Novel proteomic approaches allow the simultaneous identification of large quantities of proteins modified in an NO-dependent manner and thereby will considerably deepen our understanding of the role NO plays in cardiovascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Axel Gödecke
- Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität, Düsseldorf, Germany.
| | | | | |
Collapse
|
95
|
Bowers RR, Manevich Y, Townsend DM, Tew KD. Sulfiredoxin redox-sensitive interaction with S100A4 and non-muscle myosin IIA regulates cancer cell motility. Biochemistry 2012; 51:7740-54. [PMID: 22934964 DOI: 10.1021/bi301006w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sulfiredoxin (Srx) is a redox active protein that participates in the reduction of oxidized cysteine residues. Here we identify a novel function of Srx through its specific binding to S-glutathionylated S100A4 affecting its interaction with non-muscle myosin (NMIIA), thereby modulating the effect of S100A4 on NMIIA function and impacting cell adhesion and migration. Srx forms a complex with S100A4 (and has stronger affinity for S-glutathionylated S100A4), regulates its activity, and mediates redox regulation of the interaction of S100A4 with NMIIA. The consequence of this regulation is microfilament remodeling and altered cellular motility and adhesion. Srx-overexpressing cells had reduced levels of adhesion, decreased levels of Tyr(397)-phosphorylated focal adhesion kinase, and increased cell motility in wound healing assays. These results describe a novel redox-sensitive role for Srx in mediating complex protein interactions with plausible consequences for cancer cell motility.
Collapse
Affiliation(s)
- Robert R Bowers
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | | | | | | |
Collapse
|
96
|
Petrini S, Passarelli C, Pastore A, Tozzi G, Coccetti M, Colucci M, Bianchi M, Carrozzo R, Bertini E, Piemonte F. Protein glutathionylation in cellular compartments: a constitutive redox signal. Redox Rep 2012; 17:63-71. [PMID: 22564349 DOI: 10.1179/1351000212y.0000000009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Glutathione provides means of regulating protein function by the process of glutathionylation. Despite the role of oxidative stress biomarkers assumed recently by glutathionylated proteins in human diseases, so far no information is available on the intracellular distribution of glutathionylated proteins in human cell lines. In this study, we combined the specificity of monoclonal antibody labeling for protein-bound glutathione (GS-Pro) with the ability of confocal microscopy to localize molecules with high spatial resolution. We performed immunofluorescence analysis on dermal fibroblasts, both in steady state than in proliferative conditions, and on in situ extracted matrix samples. For the first time, we report the compartmentalization of constitutively glutathionylated proteins in different subcellular districts and we found a tight association between glutathione, nuclear lamina, and cytoskeleton. In proliferating cells, total GS-Pro fluorescence increases in the early phases of growth and significantly drops when cells reach confluence. Interestingly, a nuclear shift of GS-Pro was observed between 6 and 48 hours after plating, becoming homogeneous with the cytoplasm when growth slows. The ability to visualize a detailed intracellular distribution of this critical marker of protein oxidation may provide an additional tool to highlight pathways in turns 'redox-activated' and to identify new pathogenic pathways in human diseases.
Collapse
Affiliation(s)
- Stefania Petrini
- Laboratories of Research, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Fitzpatrick AM, Jones DP, Brown LAS. Glutathione redox control of asthma: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2012; 17:375-408. [PMID: 22304503 PMCID: PMC3353819 DOI: 10.1089/ars.2011.4198] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/22/2012] [Accepted: 01/22/2012] [Indexed: 12/11/2022]
Abstract
Asthma is a chronic inflammatory disorder of the airways associated with airway hyper-responsiveness and airflow limitation in response to specific triggers. Whereas inflammation is important for tissue regeneration and wound healing, the profound and sustained inflammatory response associated with asthma may result in airway remodeling that involves smooth muscle hypertrophy, epithelial goblet-cell hyperplasia, and permanent deposition of airway extracellular matrix proteins. Although the specific mechanisms responsible for asthma are still being unraveled, free radicals such as reactive oxygen species and reactive nitrogen species are important mediators of airway tissue damage that are increased in subjects with asthma. There is also a growing body of literature implicating disturbances in oxidation/reduction (redox) reactions and impaired antioxidant defenses as a risk factor for asthma development and asthma severity. Ultimately, these redox-related perturbations result in a vicious cycle of airway inflammation and injury that is not always amenable to current asthma therapy, particularly in cases of severe asthma. This review will discuss disruptions of redox signaling and control in asthma with a focus on the thiol, glutathione, and reduced (thiol) form (GSH). First, GSH synthesis, GSH distribution, and GSH function and homeostasis are discussed. We then review the literature related to GSH redox balance in health and asthma, with an emphasis on human studies. Finally, therapeutic opportunities to restore the GSH redox balance in subjects with asthma are discussed.
Collapse
Affiliation(s)
- Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Georgia 30322, USA.
| | | | | |
Collapse
|
98
|
Wu CL, Chou HC, Cheng CS, Li JM, Lin ST, Chen YW, Chan HL. Proteomic analysis of UVB-induced protein expression- and redox-dependent changes in skin fibroblasts using lysine- and cysteine-labeling two-dimensional difference gel electrophoresis. J Proteomics 2012; 75:1991-2014. [DOI: 10.1016/j.jprot.2011.12.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 11/27/2011] [Accepted: 12/27/2011] [Indexed: 02/02/2023]
|
99
|
Pastore A, Piemonte F. S-Glutathionylation signaling in cell biology: progress and prospects. Eur J Pharm Sci 2012; 46:279-92. [PMID: 22484331 DOI: 10.1016/j.ejps.2012.03.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 03/20/2012] [Accepted: 03/21/2012] [Indexed: 11/18/2022]
Abstract
S-Glutathionylation is a mechanism of signal transduction by which cells respond effectively and reversibly to redox inputs. The glutathionylation regulates most cellular pathways. It is involved in oxidative cellular response to insult by modulating the transcription factor Nrf2 and inducing the expression of antioxidant genes (ARE); it contributes to cell survival through nuclear translocation of NFkB and activation of survival genes, and to cell death by modulating the activity of caspase 3. It is involved in mitotic spindle formation during cell division by binding cytoskeletal proteins thus contributing to cell proliferation and differentiation. Glutathionylation also interfaces with the mechanism of phosphorylation by modulating several kinases (PKA, CK) and phosphatases (PP2A, PTEN), thus allowing a cross talk between the two processes of signal transduction. Also, skeletal RyR1 channels responsible of muscle excitation-contraction coupling appear to be sensitive to glutathionylation. Members of the ryanodine receptor super family, responsible for Ca(2) release from endoplasmic reticulum stores, contain sulfhydryl groups that function as a redox "switch", which either induces or inhibits Ca(2) release. Finally, but very importantly, glutathionylation of proteins may also act on cell metabolism by modulating enzymes involved in glycosylation, in the Krebs cycle and in mitochondrial oxidative phosphorylation. In this review, we propose a greater role for glutathionylation in cell biology: not only a cellular response to oxidative stress, but an elegant and sensitive mechanism able to respond even to subtle changes in redox balance in the different cellular compartments. Given the wide spectrum of redox-sensitive proteins, we discuss the possibility that different pathways light up by glutathionylation under various pathological conditions. The feature of reversibility of this process also makes it prone to develop targeted drug therapies and monitor the pharmacological effectiveness once identified the sensor proteins involved.
Collapse
Affiliation(s)
- Anna Pastore
- Laboratory of Biochemistry, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | |
Collapse
|
100
|
Wojtera-Kwiczor J, Groß F, Leffers HM, Kang M, Schneider M, Scheibe R. Transfer of a Redox-Signal through the Cytosol by Redox-Dependent Microcompartmentation of Glycolytic Enzymes at Mitochondria and Actin Cytoskeleton. FRONTIERS IN PLANT SCIENCE 2012; 3:284. [PMID: 23316205 PMCID: PMC3540817 DOI: 10.3389/fpls.2012.00284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/30/2012] [Indexed: 05/03/2023]
Abstract
The cytosolic glyceraldehyde-3-phosphate dehydrogenase (GAPDH, EC 1.2.1.12, GapC) plays an important role in glycolysis by providing the cell with ATP and NADH. Interestingly, despite its glycolytic function in the cytosol, GAPDH was reported to possess additional non-glycolytic activities, correlating with its nuclear, or cytoskeletal localization in animal cells. In transiently transformed mesophyll protoplasts from Arabidopsis thaliana colocalization and interaction of the glycolytic enzymes with the mitochondria and with the actin cytoskeleton was visualized by confocal laser scanning microscopy (cLSM) using fluorescent protein fusions and by bimolecular fluorescence complementation, respectively. Yeast two-hybrid screens, dot-blot overlay assays, and co-sedimentation assays were used to identify potential protein-protein interactions between two cytosolic GAPDH isoforms (GapC1, At3g04120; GapC2, At1g13440) from A. thaliana with the neighboring glycolytic enzyme, fructose 1,6-bisphosphate aldolase (FBA6, At2g36460), the mitochondrial porin (VDAC3; At5g15090), and actin in vitro. From these experiments, a mitochondrial association is suggested for both glycolytic enzymes, GAPDH and aldolase, which appear to bind to the outer mitochondrial membrane, in a redox-dependent manner. In addition, both glycolytic enzymes were found to bind to F-actin in co-sedimentation assays, and lead to bundling of purified rabbit actin, as visualized by cLSM. Actin-binding and bundling occurred reversibly under oxidizing conditions. We speculate that such dynamic formation of microcompartments is part of a redox-dependent retrograde signal transduction network for adaptation upon oxidative stress.
Collapse
Affiliation(s)
- Joanna Wojtera-Kwiczor
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
| | - Felicitas Groß
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
| | - Hans-Martin Leffers
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
| | - Minhee Kang
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
| | - Markus Schneider
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
| | - Renate Scheibe
- Department of Plant Physiology, Faculty of Biology and Chemistry, University of OsnabrueckOsnabrueck, Germany
- *Correspondence: Renate Scheibe, Department of Plant Physiology, Faculty of Biology and Chemistry, University of Osnabrueck, D-49069 Osnabrueck, Germany. e-mail:
| |
Collapse
|