101
|
Urmey AR, Zondlo NJ. Synthesis of peptides with cysteine sulfinic acid via the cysteine methoxybenzyl sulfone. Pept Sci (Hoboken) 2019. [DOI: 10.1002/pep2.24137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Andrew R. Urmey
- Department of Chemistry and Biochemistry University of Delaware Newark Delaware United States
| | - Neal J. Zondlo
- Department of Chemistry and Biochemistry University of Delaware Newark Delaware United States
| |
Collapse
|
102
|
Kolupaev YE, Karpets YV, Kabashnikova LF. Antioxidative System of Plants: Cellular Compartmentalization, Protective and Signaling Functions, Mechanisms of Regulation (Review). APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s0003683819050089] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
103
|
Zeida A, Trujillo M, Ferrer-Sueta G, Denicola A, Estrin DA, Radi R. Catalysis of Peroxide Reduction by Fast Reacting Protein Thiols. Chem Rev 2019; 119:10829-10855. [PMID: 31498605 DOI: 10.1021/acs.chemrev.9b00371] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Life on Earth evolved in the presence of hydrogen peroxide, and other peroxides also emerged before and with the rise of aerobic metabolism. They were considered only as toxic byproducts for many years. Nowadays, peroxides are also regarded as metabolic products that play essential physiological cellular roles. Organisms have developed efficient mechanisms to metabolize peroxides, mostly based on two kinds of redox chemistry, catalases/peroxidases that depend on the heme prosthetic group to afford peroxide reduction and thiol-based peroxidases that support their redox activities on specialized fast reacting cysteine/selenocysteine (Cys/Sec) residues. Among the last group, glutathione peroxidases (GPxs) and peroxiredoxins (Prxs) are the most widespread and abundant families, and they are the leitmotif of this review. After presenting the properties and roles of different peroxides in biology, we discuss the chemical mechanisms of peroxide reduction by low molecular weight thiols, Prxs, GPxs, and other thiol-based peroxidases. Special attention is paid to the catalytic properties of Prxs and also to the importance and comparative outlook of the properties of Sec and its role in GPxs. To finish, we describe and discuss the current views on the activities of thiol-based peroxidases in peroxide-mediated redox signaling processes.
Collapse
Affiliation(s)
| | | | | | | | - Darío A Estrin
- Departamento de Química Inorgánica, Analítica y Química-Física and INQUIMAE-CONICET , Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , 2160 Buenos Aires , Argentina
| | | |
Collapse
|
104
|
Hepatitis C Virus RNA-Dependent RNA Polymerase Is Regulated by Cysteine S-Glutathionylation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3196140. [PMID: 31687077 PMCID: PMC6800943 DOI: 10.1155/2019/3196140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022]
Abstract
Hepatitis C virus (HCV) triggers massive production of reactive oxygen species (ROS) and affects expression of genes encoding ROS-scavenging enzymes. Multiple lines of evidence show that levels of ROS production contribute to the development of various virus-associated pathologies. However, investigation of HCV redox biology so far remained in the paradigm of oxidative stress, whereas no attention was given to the identification of redox switches among viral proteins. Here, we report that one of such redox switches is the NS5B protein that exhibits RNA-dependent RNA polymerase (RdRp) activity. Treatment of the recombinant protein with reducing agents significantly increases its enzymatic activity. Moreover, we show that the NS5B protein is subjected to S-glutathionylation that affects cysteine residues 89, 140, 170, 223, 274, 521, and either 279 or 295. Substitution of these cysteines except C89 and C223 with serine residues led to the reduction of the RdRp activity of the recombinant protein in a primer-dependent assay. The recombinant protein with a C279S mutation was almost inactive in vitro and could not be activated with reducing agents. In contrast, cysteine substitutions in the NS5B region in the context of a subgenomic replicon displayed opposite effects: most of the mutations enhanced HCV replication. This difference may be explained by the deleterious effect of oxidation of NS5B cysteine residues in liver cells and by the protective role of S-glutathionylation. Based on these data, redox-sensitive posttranslational modifications of HCV NS5B and other proteins merit a more detailed investigation and analysis of their role(s) in the virus life cycle and associated pathogenesis.
Collapse
|
105
|
Cobley JN, Noble A, Jimenez-Fernandez E, Valdivia Moya MT, Guille M, Husi H. Catalyst-free Click PEGylation reveals substantial mitochondrial ATP synthase sub-unit alpha oxidation before and after fertilisation. Redox Biol 2019; 26:101258. [PMID: 31234016 PMCID: PMC6597785 DOI: 10.1016/j.redox.2019.101258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/21/2022] Open
Abstract
Using non-reducing Western blotting to assess protein thiol redox state is challenging because most reduced and oxidised forms migrate at the same molecular weight and are, therefore, indistinguishable. While copper catalysed Click chemistry can be used to ligate a polyethylene glycol (PEG) moiety termed Click PEGylation to mass shift the reduced or oxidised form as desired, the potential for copper catalysed auto-oxidation is problematic. Here we define a catalyst-free trans-cyclooctene-methyltetrazine (TCO-Tz) inverse electron demand Diels Alder chemistry approach that affords rapid (k ~2000 M-1 s-1), selective and bio-orthogonal Click PEGylation. We used TCO-Tz Click PEGylation to investigate how fertilisation impacts reversible mitochondrial ATP synthase F1-Fo sub-unit alpha (ATP-α-F1) oxidation-an established molecular correlate of impaired enzyme activity-in Xenopus laevis. TCO-Tz Click PEGylation studies reveal substantial (~65%) reversible ATP-α-F1 oxidation at evolutionary conserved cysteine residues (i.e., C244 and C294) before and after fertilisation. A single thiol is, however, preferentially oxidised likely due to greater solvent exposure during the catalytic cycle. Selective reduction experiments show that: S-glutathionylation accounts for ~50-60% of the reversible oxidation observed, making it the dominant oxidative modification type. Intermolecular disulphide bonds may also contribute due to their relative stability. Substantial reversible ATP-α-F1 oxidation before and after fertilisation is biologically meaningful because it implies low mitochondrial F1-Fo ATP synthase activity. Catalyst-free TCO-Tz Click PEGylation is a valuable new tool to interrogate protein thiol redox state in health and disease.
Collapse
Affiliation(s)
- James N Cobley
- Free Radical Research Group, University of the Highlands and Islands, Centre for Health Sciences, Inverness, IV2 3JH, UK.
| | - Anna Noble
- European Xenopus Resource Centre, University of Portsmouth, School of Biological Sciences, King Henry Building, Portsmouth, PO1 2DY, UK
| | - Eduardo Jimenez-Fernandez
- Free Radical Research Group, University of the Highlands and Islands, Centre for Health Sciences, Inverness, IV2 3JH, UK
| | - Manuel-Thomas Valdivia Moya
- Free Radical Research Group, University of the Highlands and Islands, Centre for Health Sciences, Inverness, IV2 3JH, UK
| | - Matthew Guille
- European Xenopus Resource Centre, University of Portsmouth, School of Biological Sciences, King Henry Building, Portsmouth, PO1 2DY, UK
| | - Holger Husi
- Free Radical Research Group, University of the Highlands and Islands, Centre for Health Sciences, Inverness, IV2 3JH, UK
| |
Collapse
|
106
|
Alli-Oluwafuyi AM, Luis PB, Nakashima F, Giménez-Bastida JA, Presley SH, Duvernay MT, Iwalewa EO, Schneider C. Curcumin induces secretion of glucagon-like peptide-1 through an oxidation-dependent mechanism. Biochimie 2019; 165:250-257. [PMID: 31470039 DOI: 10.1016/j.biochi.2019.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/24/2019] [Indexed: 10/26/2022]
Abstract
Curcumin shows antiglycemic effects in animals. Curcumin is chemically unstable at physiological pH, and its oxidative degradation products were shown to contribute to its anti-inflammatory effects. Since the degradation products may also contribute to other effects, we analyzed their role in the antiglycemic activity of curcumin. We quantified curcumin-induced release of glucagon-like peptide 1 (GLP-1) from mouse STC-1 cells that represent enteroendocrine L-cells as a major source of this anti-diabetic hormone. Curcumin induced secretion of GLP-1 in a dose-dependent manner. Two chemically stable analogues of curcumin that do not readily undergo degradation, were less active while two unstable analogues were active secretagogues. Chromatographically isolated spiroepoxide, an unstable oxidative metabolite of curcumin with anti-inflammatory activity, also induced secretion of GLP-1. Stable compounds like the final oxidative metabolite bicyclopentadione, and the major plasma metabolite, curcumin-glucuronide, were inactive. GLP-1 secretion induced by curcumin and its oxidative degradation products was associated with activation of PKC, ERK, and CaM kinase II. Since activity largely correlated with instability of curcumin and the analogues, we tested the extent of covalent binding to proteins in STC-1 cells and found it occurred with similar affinity as N-ethylmaleimide, indicating covalent binding occurred with nucleophilic cysteine residues. These results suggest that oxidative metabolites of curcumin are involved in the antiglycemic effects of curcumin. Our findings support the hypothesis that curcumin functions as a pro-drug requiring oxidative activation to reveal its bioactive metabolites that act by binding to target proteins thereby causing a change in function.
Collapse
Affiliation(s)
- Abdul-Musawwir Alli-Oluwafuyi
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA; Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria
| | - Paula B Luis
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Fumie Nakashima
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Juan A Giménez-Bastida
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Sai Han Presley
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Matthew T Duvernay
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA
| | - Ezekiel O Iwalewa
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Claus Schneider
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, 37232, USA.
| |
Collapse
|
107
|
Olson KR. Hydrogen sulfide, reactive sulfur species and coping with reactive oxygen species. Free Radic Biol Med 2019; 140:74-83. [PMID: 30703482 DOI: 10.1016/j.freeradbiomed.2019.01.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/19/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022]
Abstract
Life began in a ferruginous (anoxic and Fe2+ dominated) world around 3.8 billion years ago (bya). Hydrogen sulfide (H2S) and other sulfur molecules from hydrothermal vents and other fissures provided many key necessities for life's origin including catalytic platforms (primordial enzymes) that also served as primitive boundaries (cell walls), substrates for organic synthesis and a continuous source of energy in the form of reducing equivalents. Anoxigenic photosynthesis oxidizing H2S followed within a few hundred million years and laid the metabolic groundwork for oxidative photosynthesis some half-billion years later that slightly and episodically increased atmospheric oxygen around 2.3 bya. This oxidized terrestrial sulfur to sulfate which was washed to the sea where it was reduced creating vast euxinic (anoxic and sulfidic) areas. It was in this environment that eukaryotic cells appeared around 1.5 bya and where they evolved for nearly 1 billion additional years. Oxidative photosynthesis finally oxidized the oceans and around 0.6 bya oxygen levels in the atmosphere and oceans began to rise toward present day levels. This is purported to have been a life-threatening event due to the prevalence of reactive oxygen species (ROS) and thus necessitated the elaboration of chemical and enzymatic antioxidant mechanisms. However, these antioxidants initially appeared around the time of anoxigenic photosynthesis suggesting a commitment to metabolism of reactive sulfur species (RSS). This review examines these events and suggests that many of the biological attributes assigned to ROS may, in fact, be due to RSS. This is underscored by observations that ROS and RSS are chemically similar, often indistinguishable by analytical methods and the fact that the bulk of biochemical and physiological experiments are performed in unphysiologically oxic environments where ROS are artifactually favored over RSS.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine-South Bend, Raclin Carmichael Hall, 1234 Notre Dame Ave, South Bend, IN 46617, USA.
| |
Collapse
|
108
|
Vukelic S, Xu Q, Seidel-Rogol B, Faidley EA, Dikalova AE, Hilenski LL, Jorde U, Poole LB, Lassègue B, Zhang G, Griendling KK. NOX4 (NADPH Oxidase 4) and Poldip2 (Polymerase δ-Interacting Protein 2) Induce Filamentous Actin Oxidation and Promote Its Interaction With Vinculin During Integrin-Mediated Cell Adhesion. Arterioscler Thromb Vasc Biol 2019; 38:2423-2434. [PMID: 30354218 DOI: 10.1161/atvbaha.118.311668] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective- Actin cytoskeleton assembly and organization, as a result of focal adhesion (FA) formation during cell adhesion, are dependent on reactive oxygen species and the cellular redox environment. Poldip2 (polymerase δ-interacting protein 2), a novel regulator of NOX4 (NADPH oxidase 4), plays a significant role in reactive oxygen species production and cytoskeletal remodeling. Thus, we hypothesized that endogenous reactive oxygen species derived from Poldip2/NOX4 contribute to redox regulation of actin and cytoskeleton assembly during integrin-mediated cell adhesion. Approach and Results- Using vascular smooth muscle cells, we verified that hydrogen peroxide (H2O2) levels increase during integrin-mediated cell attachment as a result of activation of NOX4. Filamentous actin (F-actin) was oxidized by sulfenylation during cell attachment, with a peak at 3 hours (0.80±0.04 versus 0.08±0.13 arbitrary units at time zero), which was enhanced by overexpression of Poldip2. Depletion of Poldip2 or NOX4 using siRNA, or scavenging of endogenous H2O2 with catalase, inhibited F-actin oxidation by 78±26%, 99±1%, and 98±1%, respectively. To determine the consequence of F-actin oxidation, we examined the binding of F-actin to vinculin, a protein involved in FA complexes that regulates FA maturation. Vinculin binding during cell adhesion as well as migration capacity were inhibited after transfection with actin containing 2 oxidation-resistant point mutations (C272A and C374A). Silencing of Poldip2 or NOX4 also impaired actin-vinculin interaction, which disturbed maturation of FAs and inhibited cell migration. Conclusions- These results suggest that integrin engagement during cell attachment activates Poldip2/Nox4 to oxidize actin, which modulates FA assembly.
Collapse
Affiliation(s)
- Sasa Vukelic
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.).,Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (S.V., U.J.)
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.).,Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (Q.X., G.Z.)
| | - Bonnie Seidel-Rogol
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Elizabeth A Faidley
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Anna E Dikalova
- Department of Medicine, Vanderbilt University, Nashville, TN (A.E.D.)
| | - Lula L Hilenski
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Ulrich Jorde
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (S.V., U.J.)
| | - Leslie B Poole
- Department of Biochemistry, Center for Molecular Signaling, Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, NC (L.B.P.)
| | - Bernard Lassègue
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (Q.X., G.Z.)
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| |
Collapse
|
109
|
Batinic-Haberle I, Tome ME. Thiol regulation by Mn porphyrins, commonly known as SOD mimics. Redox Biol 2019; 25:101139. [PMID: 31126869 PMCID: PMC6859569 DOI: 10.1016/j.redox.2019.101139] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/18/2019] [Accepted: 02/07/2019] [Indexed: 01/27/2023] Open
Abstract
Superoxide dismutases play an important role in human health and disease. Three decades of effort have gone into synthesizing SOD mimics for clinical use. The result is the Mn porphyrins which have SOD-like activity. Several clinical trials are underway to test the efficacy of these compounds in patients, particularly as radioprotectors of normal tissue during cancer treatment. However, aqueous chemistry data indicate that the Mn porphyrins react equally well with multiple redox active species in cells including H2O2, O2•-, ONOO-, thiols, and ascorbate among others. The redox potential of the Mn porphyrins is midway between the potentials for the oxidation and reduction of O2•-. This positions them to react equally well as oxidants and reductants in cells. The result of this unique chemistry is that: 1) the species the Mn porphyrins react with in vivo will depend on the relative concentrations of the reactive species and Mn porphyrins in the cell of interest, and 2) the Mn porphyrins will act as catalytic (redox cycling) agents in vivo. The ability of the Mn porphyrins to catalyze protein S-glutathionylation means that Mn porphyrins have the potential to globally modulate cellular redox regulatory signaling networks. The purpose of this review is to summarize the data that indicate the Mn porphyrins have diverse reactions in vivo that are the basis of the observed biological effects. The ability to catalyze multiple reactions in vivo expands the potential therapeutic use of the Mn porphyrins to disease models that are not SOD based.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Margaret E Tome
- Departments of Pathology and Pharmacology, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
110
|
Ferguson GD, Bridge WJ. The glutathione system and the related thiol network in Caenorhabditis elegans. Redox Biol 2019. [DOI: 10.1110.1016/j.redox.2019.101171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
111
|
AGE-RAGE stress: a changing landscape in pathology and treatment of Alzheimer's disease. Mol Cell Biochem 2019; 459:95-112. [PMID: 31079281 DOI: 10.1007/s11010-019-03553-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022]
Abstract
Numerous hypotheses including amyloid cascade, cholinergic, and oxidative have been proposed for pathogenesis of Alzheimer's disease (AD). The data suggest that advanced glycation end products (AGEs) and its receptor RAGE (receptor for AGE) are involved in the pathogenesis of AD. AGE-RAGE stress, defined as a balance between stressors (AGE, RAGE) and anti-stressors (sRAGE, AGE degraders) in favor of stressors, has been implicated in pathogenesis of diseases. AGE and its interaction with RAGE-mediated increase in the reactive oxygen species (ROS) damage brain because of its increased vulnerability to ROS. AGE and ROS increase the synthesis of amyloid β (Aβ) leading to deposition of Aβ and phosphorylation of tau, culminating in formation of plaques and neurofibrillary tangles. ROS increase the synthesis of Aβ, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Elevation of ROS precedes the Aβ plaques formation. Because of involvement of AGE and RAGE in AD pathology, the treatment should be targeted at lowering AGE levels through reduction in consumption and formation of AGE, and lowering expression of RAGE, blocking of RAGE ligand binding, increasing levels of soluble RAGE (sRAGE), and use of antioxidants. The above treatment aspect of AD is lacking. In conclusion, AGE-RAGE stress initiates, and Aβ, HMGB1, and S100 enhance the progression of AD. Reduction of levels of AGE and RAGE, elevation of sRAGE, and antioxidants would be beneficial therapeutic modalities in the prevention, regression, and slowing of progression of AD.
Collapse
|
112
|
The science of licking your wounds: Function of oxidants in the innate immune system. Biochem Pharmacol 2019; 163:451-457. [DOI: 10.1016/j.bcp.2019.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/08/2019] [Indexed: 02/07/2023]
|
113
|
Olson KR, Gao Y. Effects of inhibiting antioxidant pathways on cellular hydrogen sulfide and polysulfide metabolism. Free Radic Biol Med 2019; 135:1-14. [PMID: 30790656 DOI: 10.1016/j.freeradbiomed.2019.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022]
Abstract
Elaborate antioxidant pathways have evolved to minimize the threat of excessive reactive oxygen species (ROS) and to regulate ROS as signaling entities. ROS are chemically and functionally similar to reactive sulfur species (RSS) and both ROS and RSS have been shown to be metabolized by the antioxidant enzymes, superoxide dismutase and catalase. Here we use fluorophores to examine the effects of a variety of inhibitors of antioxidant pathways on metabolism of two important RSS, hydrogen sulfide (H2S with AzMC) and polysulfides (H2Sn, where n = 2-7, with SSP4) in HEK293 cells. Cells were exposed to inhibitors for up to 5 days in normoxia (21% O2) and hypoxia (5% O2), conditions also known to affect ROS production. Decreasing intracellular glutathione (GSH) with l-buthionine-sulfoximine (BSO) or diethyl maleate (DEM) decreased H2S production for 5 days but did not affect H2Sn. The glutathione reductase inhibitor, auranofin, initially decreased H2S and H2Sn but after two days H2Sn increased over controls. Inhibition of peroxiredoxins with conoidin A decreased H2S and increased H2Sn, whereas the glutathione peroxidase inhibitor, tiopronin, increased H2S. Aminoadipic acid, an inhibitor of cystine uptake did not affect either H2S or H2Sn. In buffer, the glutathione reductase and thioredoxin reductase inhibitor, 2-AAPA, the glutathione peroxidase mimetic, ebselen, and tiopronin variously reacted directly with AzMC and SSP4, reacted with H2S and H2S2, or optically interfered with AzMC or SSP4 fluorescence. Collectively these results show that antioxidant inhibitors, generally known for their ability to increase cellular ROS, have various effects on cellular RSS. These findings suggest that the inhibitors may affect cellular sulfur metabolism pathways that are not related to ROS production and in some instances they may directly affect RSS or the methods used to measure them. They also illustrate the importance of carefully evaluating RSS metabolism when biologically or pharmacologically attempting to manipulate ROS.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine - South Bend, South Bend, IN, 46617, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Yan Gao
- Indiana University School of Medicine - South Bend, South Bend, IN, 46617, USA
| |
Collapse
|
114
|
Structure and mechanisms of ROS generation by NADPH oxidases. Curr Opin Struct Biol 2019; 59:91-97. [PMID: 31051297 DOI: 10.1016/j.sbi.2019.03.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/14/2019] [Accepted: 03/04/2019] [Indexed: 12/31/2022]
Abstract
NADPH oxidases (NOXs) are integral membrane enzymes that produce reactive oxygen species. Humans have seven NOX enzymes that feature a very similar catalytic core but distinct regulatory mechanisms. The recent structural elucidation of the NOX catalytic domains has been a step forward in the field. NADPH, FAD, and two hemes form a linear array of redox cofactors that transfer electrons across to the two sides of the membrane. Oxygen is reduced through an unusual outer sphere mechanism that does not involve any covalent intermediate with the heme iron. Several recent studies have expanded the roles of NOXs in cell signaling, innate immune response, and cell proliferation including oncogenic transformation. This work reinforces NOX-generated ROS as powerful signaling molecules. A challenging question is to understand the specific mechanisms of enzyme regulation and to harness the growing insight on NOXs' structure and biochemistry to generate more powerful small-molecule modulators of NOX activities.
Collapse
|
115
|
Guerra-Moreno A, Ang J, Welsch H, Jochem M, Hanna J. Regulation of the unfolded protein response in yeast by oxidative stress. FEBS Lett 2019; 593:1080-1088. [PMID: 31002390 DOI: 10.1002/1873-3468.13389] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
In the unfolded protein response (UPR), Ire1 activates Hac1 to coordinate the transcription of hundreds of genes to mitigate ER stress. Recent work in Caenorhabditis elegans suggests that oxidative stress inhibits this canonical Ire1 signalling pathway, activating instead an antioxidant stress response. We sought to determine whether this novel mode of UPR function also existed in yeast, where Ire1 has been best characterized. We show that the yeast UPR is also subject to inhibition by oxidative stress. Inhibition is mediated by a single evolutionarily conserved cysteine, and affects both luminal and membrane pathways of Ire1 activation. In yeast, Ire1 appears dispensable for resistance to oxidative stress and, therefore, the physiological significance of this pathway remains to be demonstrated.
Collapse
Affiliation(s)
- Angel Guerra-Moreno
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessie Ang
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hendrik Welsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marco Jochem
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - John Hanna
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
116
|
Dissmeyer N. Conditional Protein Function via N-Degron Pathway-Mediated Proteostasis in Stress Physiology. ANNUAL REVIEW OF PLANT BIOLOGY 2019; 70:83-117. [PMID: 30892918 DOI: 10.1146/annurev-arplant-050718-095937] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The N-degron pathway, formerly the N-end rule pathway, regulates functions of regulatory proteins. It impacts protein half-life and therefore directs the actual presence of target proteins in the cell. The current concept holds that the N-degron pathway depends on the identity of the amino (N)-terminal amino acid and many other factors, such as the follow-up sequence at the N terminus, conformation, flexibility, and protein localization. It is evolutionarily conserved throughout the kingdoms. One possible entry point for substrates of the N-degron pathway is oxidation of N-terminal Cys residues. Oxidation of N-terminal Cys is decisive for further enzymatic modification of various neo-N termini by arginylation that generates potentially neofunctionalized or instable proteoforms. Here, I focus on the posttranslational modifications that are encompassed by protein degradation via the Cys/Arg branch of the N-degron pathway-part of the PROTEOLYSIS 6 (PRT6)/N-degron pathway-as well as the underlying physiological principles of this branch and its biological significance in stress response.
Collapse
Affiliation(s)
- Nico Dissmeyer
- Independent Junior Research Group on Protein Recognition and Degradation, Leibniz Institute of Plant Biochemistry (IPB) and ScienceCampus Halle-Plant-Based Bioeconomy, D-06120 Halle (Saale), Germany; ; Twitter: @NDissmeyer
| |
Collapse
|
117
|
Abstract
SIGNIFICANCE Cellular reactive oxygen species (ROS) mediate redox signaling cascades that are critical to numerous physiological and pathological processes. Analytical methods to monitor cellular ROS levels and proteomic platforms to identify oxidative post-translational modifications (PTMs) of proteins are critical to understanding the triggers and consequences of redox signaling. Recent Advances: The prevalence and significance of redox signaling has recently been illuminated through the use of chemical probes that allow for sensitive detection of cellular ROS levels and proteomic dissection of oxidative PTMs directly in living cells. CRITICAL ISSUES In this review, we provide a comprehensive overview of chemical probes that are available for monitoring ROS and oxidative PTMs, and we highlight the advantages and limitations of these methods. FUTURE DIRECTIONS Despite significant advances in chemical probes, the low levels of cellular ROS and low stoichiometry of oxidative PTMs present challenges for accurately measuring the extent and dynamics of ROS generation and redox signaling. Further improvements in sensitivity and ability to spatially and temporally control readouts are essential to fully illuminate cellular redox signaling.
Collapse
Affiliation(s)
- Masahiro Abo
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts
| | | |
Collapse
|
118
|
Spiers JG, Chen HJC, Bourgognon JM, Steinert JR. Dysregulation of stress systems and nitric oxide signaling underlies neuronal dysfunction in Alzheimer's disease. Free Radic Biol Med 2019; 134:468-483. [PMID: 30716433 DOI: 10.1016/j.freeradbiomed.2019.01.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022]
Abstract
Stress is a multimodal response involving the coordination of numerous body systems in order to maximize the chance of survival. However, long term activation of the stress response results in neuronal oxidative stress via reactive oxygen and nitrogen species generation, contributing to the development of depression. Stress-induced depression shares a high comorbidity with other neurological conditions including Alzheimer's disease (AD) and dementia, often appearing as one of the earliest observable symptoms in these diseases. Furthermore, stress and/or depression appear to exacerbate cognitive impairment in the context of AD associated with dysfunctional catecholaminergic signaling. Given there are a number of homologous pathways involved in the pathophysiology of depression and AD, this article will highlight the mechanisms by which stress-induced perturbations in oxidative stress, and particularly NO signaling, contribute to neurodegeneration.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, 3083, Australia.
| | - Hsiao-Jou Cortina Chen
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | | | - Joern R Steinert
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 9HN, United Kingdom.
| |
Collapse
|
119
|
McConnell EW, Berg P, Westlake TJ, Wilson KM, Popescu GV, Hicks LM, Popescu SC. Proteome-Wide Analysis of Cysteine Reactivity during Effector-Triggered Immunity. PLANT PHYSIOLOGY 2019; 179:1248-1264. [PMID: 30510037 PMCID: PMC6446758 DOI: 10.1104/pp.18.01194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 05/08/2023]
Abstract
A surge in the accumulation of oxidants generates shifts in the cellular redox potential during early stages of plant infection with pathogens and activation of effector-triggered immunity (ETI). The redoxome, defined as the proteome-wide oxidative modifications of proteins caused by oxidants, has a well-known impact on stress responses in metazoans. However, the identity of proteins and the residues sensitive to oxidation during the plant immune response remain largely unknown. Previous studies of the thimet oligopeptidases TOP1 and TOP2 placed them in the salicylic acid dependent branch of ETI, with a current model wherein TOPs sustain interconnected organellar and cytosolic pathways that modulate the oxidative burst and development of cell death. Herein, we characterized the ETI redoxomes in Arabidopsis (Arabidopsis thaliana) wild-type Col-0 and top1top2 mutant plants using a differential alkylation-based enrichment technique coupled with label-free mass spectrometry-based quantification. We identified cysteines sensitive to oxidation in a wide range of protein families at multiple time points after pathogen infection. Differences were detected between Col-0 and top1top2 redoxomes regarding the identity and number of oxidized cysteines, and the amplitude of time-dependent fluctuations in protein oxidation. Our results support a determining role for TOPs in maintaining the proper level and dynamics of proteome oxidation during ETI. This study significantly expands the repertoire of oxidation-sensitive plant proteins and can guide future mechanistic studies.
Collapse
Affiliation(s)
- Evan W McConnell
- Department of Chemistry, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Philip Berg
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, Mississippi State, Mississippi 39762
| | - Timothy J Westlake
- Department of Plant Pathology and Plant-Microbe Biology, Cornell University, Ithaca, New York 14850
| | - Katherine M Wilson
- Department of Chemistry, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - George V Popescu
- Institute for Genomics, Biocomputing and Biotechnology, Mississippi State University, Mississippi State, Mississippi 39762
- The National Institute for Laser, Plasma & Radiation Physics, 077126 Măgurele, Ilfov, Romania
| | - Leslie M Hicks
- Department of Chemistry, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Sorina C Popescu
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, Mississippi State, Mississippi 39762
| |
Collapse
|
120
|
Ferguson GD, Bridge WJ. The glutathione system and the related thiol network in Caenorhabditis elegans. Redox Biol 2019; 24:101171. [PMID: 30901603 PMCID: PMC6429583 DOI: 10.1016/j.redox.2019.101171] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 01/09/2023] Open
Abstract
Advances in the field of redox biology have contributed to the understanding of the complexity of the thiol-based system in mediating signal transduction. The redox environment is the overall spatiotemporal balance of oxidation-reduction systems within the integrated compartments of the cell, tissues and whole organisms. The ratio of the reduced to disulfide glutathione redox couple (GSH:GSSG) is a key indicator of the redox environment and its associated cellular health. The reaction mechanisms of glutathione-dependent and related thiol-based enzymes play a fundamental role in the function of GSH as a redox regulator. Glutathione homeostasis is maintained by the balance of GSH synthesis (de novo and salvage pathways) and its utilization through its detoxification, thiol signalling, and antioxidant defence functions via GSH-dependent enzymes and free radical scavenging. As such, GSH acts in concert with the entire redox network to maintain reducing conditions in the cell. Caenorhabditis elegans offers a simple model to facilitate further understanding at the multicellular level of the physiological functions of GSH and the GSH-dependent redox network. This review discusses the C. elegans studies that have investigated glutathione and related systems of the redox network including; orthologs to the protein-encoding genes of GSH synthesis; glutathione peroxidases; glutathione-S-transferases; and the glutaredoxin, thioredoxin and peroxiredoxin systems.
Collapse
Affiliation(s)
- Gavin Douglas Ferguson
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Wallace John Bridge
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
121
|
Wang J. Crystallographic identification of spontaneous oxidation intermediates and products of protein sulfhydryl groups. Protein Sci 2019; 28:472-477. [PMID: 30592103 PMCID: PMC6371210 DOI: 10.1002/pro.3568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/23/2018] [Accepted: 12/26/2018] [Indexed: 11/12/2022]
Abstract
In the absence of protective reducing agents, Cys residues in purified proteins can be oxidized spontaneously by oxygen in the air, as frequently observed in protein crystal structures. However, the formation of an O-bridge via dehydration mechanism between a peroxidized Cys side chain and a primary amine of Lys side chain in proteins has not yet been reported. When an electron density feature was observed for an extra group or an extra atom between side chains of Cys-245 and Lys-158 in the crystal structure of histidinol phosphate phosphatase, mass spectrometric analysis was carried out for its chemical identification. That analysis led to a conclusion that this extra density corresponded to a methylene group. It was then proposed that these two residues were able to absorb CO2 and reduced it to CH2 spontaneously. Further examination of other protein structures in the PDB showed that the formation of this cross-linking species was a widespread phenomenon. This claim is examined in this study using methods recently developed for quantification of electrons around nucleus as the means for direct chemical identification. It is found that an O-bridge is actually formed between Cys and Lys side chains, instead of a CH2 -bridge.
Collapse
Affiliation(s)
- Jimin Wang
- Department of Molecular Biophysics and BiochemistryYale UniversityNew HavenConnecticut06520
| |
Collapse
|
122
|
A battery of translational biomarkers for the assessment of the in vitro and in vivo antioxidant action of plant polyphenolic compounds: The biomarker issue. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2018.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
123
|
Zhu X, Meyers A, Long D, Ingram B, Liu T, Yoza BK, Vachharajani V, McCall CE. Frontline Science: Monocytes sequentially rewire metabolism and bioenergetics during an acute inflammatory response. J Leukoc Biol 2019; 105:215-228. [PMID: 30633362 DOI: 10.1002/jlb.3hi0918-373r] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/26/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolism directs the severe acute inflammatory reaction of monocytes to guard homeostasis. This occurs by sequentially activating anabolic immune effector mechanisms, switching to immune deactivation mechanisms and then restoring immunometabolic homeostasis. Nuclear sirtuin 1 and mitochondrial pyruvate dehydrogenase kinase metabolically drive this dynamic and are druggable targets that promote immunometabolic resolution in septic mice and increase survival. We used unbiased metabolomics and a validated monocyte culture model of activation, deactivation, and partial resolution of acute inflammation to sequentially track metabolic rewiring. Increases in glycogenolysis, hexosamine, glycolysis, and pentose phosphate pathways were aligned with anabolic activation. Activation transitioned to combined lipid, protein, amino acid, and nucleotide catabolism during deactivation, and partially subsided during early resolution. Lipid metabolic rewiring signatures aligned with deactivation included elevated n-3 and n-6 polyunsaturated fatty acids and increased levels of fatty acid acylcarnitines. Increased methionine to homocysteine cycling increased levels of s-adenosylmethionine rate-limiting transmethylation mediator, and homocysteine and cysteine transsulfuration preceded increases in glutathione. Increased tryptophan catabolism led to elevated kynurenine and de novo biosynthesis of nicotinamide adenine dinucleotide from quinolinic acid. Increased branched-chain amino acid catabolism paralleled increases in succinyl-CoA. A rise in the Krebs cycle cis-aconitate-derived itaconate and succinate with decreased fumarate and acetyl-CoA levels occurred concomitant with deactivation and subsided during early resolution. The data suggest that rewiring of metabolic and mitochondrial bioenergetics by monocytes sequentially activates, deactivates, and resolves acute inflammation.
Collapse
Affiliation(s)
- Xuewei Zhu
- Department of Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Allison Meyers
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David Long
- Department of Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Brian Ingram
- Metabolon, Inc., Morrisville, North Carolina, USA
| | - Tiefu Liu
- Department of Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Barbara K Yoza
- Department of Surgery/General Surgery and Trauma, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Vidula Vachharajani
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Charles E McCall
- Department of Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
124
|
Reichmann D, Voth W, Jakob U. Maintaining a Healthy Proteome during Oxidative Stress. Mol Cell 2019; 69:203-213. [PMID: 29351842 DOI: 10.1016/j.molcel.2017.12.021] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/11/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022]
Abstract
Some of the most challenging stress conditions that organisms encounter during their lifetime involve the transient accumulation of reactive oxygen and chlorine species. Extremely reactive to amino acid side chains, these oxidants cause widespread protein unfolding and aggregation. It is therefore not surprising that cells draw on a variety of different strategies to counteract the damage and maintain a healthy proteome. Orchestrated largely by direct changes in the thiol oxidation status of key proteins, the response strategies involve all layers of protein protection. Reprogramming of basic biological functions helps decrease nascent protein synthesis and restore redox homeostasis. Mobilization of oxidative stress-activated chaperones and production of stress-resistant non-proteinaceous chaperones prevent irreversible protein aggregation. Finally, redox-controlled increase in proteasome activity removes any irreversibly damaged proteins. Together, these systems pave the way to restore protein homeostasis and enable organisms to survive stress conditions that are inevitable when living an aerobic lifestyle.
Collapse
Affiliation(s)
- Dana Reichmann
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| | - Wilhelm Voth
- Department of Molecular, Cellular, and Developmental Biology and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Ursula Jakob
- Department of Molecular, Cellular, and Developmental Biology and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
125
|
Mulkidjanian AY, Shalaeva DN, Lyamzaev KG, Chernyak BV. Does Oxidation of Mitochondrial Cardiolipin Trigger a Chain of Antiapoptotic Reactions? BIOCHEMISTRY (MOSCOW) 2018; 83:1263-1278. [PMID: 30472963 DOI: 10.1134/s0006297918100115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Oxidative stress causes selective oxidation of cardiolipin (CL), a four-tail lipid specific for the inner mitochondrial membrane. Interaction with oxidized CL transforms cytochrome c into peroxidase capable of oxidizing even more CL molecules. Ultimately, this chain of events leads to the pore formation in the outer mitochondrial membrane and release of mitochondrial proteins, including cytochrome c, into the cytoplasm. In the cytoplasm, cytochrome c promotes apoptosome assembly that triggers apoptosis (programmed cell death). Because of this amplification cascade, even an occasional oxidation of a single CL molecule by endogenously formed reactive oxygen species (ROS) might cause cell death, unless the same CL oxidation triggers a separate chain of antiapoptotic reactions that would prevent the CL-mediated apoptotic cascade. Here, we argue that the key function of CL in mitochondria and other coupling membranes is to prevent proton leak along the interface of interacting membrane proteins. Therefore, CL oxidation should increase proton permeability through the CL-rich clusters of membrane proteins (CL islands) and cause a drop in the mitochondrial membrane potential (MMP). On one hand, the MMP drop should hinder ROS generation and further CL oxidation in the entire mitochondrion. On the other hand, it is known to cause rapid fission of the mitochondrial network and formation of many small mitochondria, only some of which would contain oxidized CL islands. The fission of mitochondrial network would hinder apoptosome formation by preventing cytochrome c release from healthy mitochondria, so that slowly working protein quality control mechanisms would have enough time to eliminate mitochondria with the oxidized CL. Because of these two oppositely directed regulatory pathways, both triggered by CL oxidation, the fate of the cell appears to be determined by the balance between the CL-mediated proapoptotic and antiapoptotic reactions. Since this balance depends on the extent of CL oxidation, mitochondria-targeted antioxidants might be able to ensure cell survival in many pathologies by preventing CL oxidation.
Collapse
Affiliation(s)
- A Y Mulkidjanian
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia. .,Lomonosov Moscow State University, School of Bioengineering and Bioinformatics, Moscow, 119991, Russia.,Osnabrueck University, Department of Physics, 49069 Osnabrueck, Germany
| | - D N Shalaeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - K G Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - B V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
126
|
Chen X, Mims J, Huang X, Singh N, Motea E, Planchon SM, Beg M, Tsang AW, Porosnicu M, Kemp ML, Boothman DA, Furdui CM. Modulators of Redox Metabolism in Head and Neck Cancer. Antioxid Redox Signal 2018; 29:1660-1690. [PMID: 29113454 PMCID: PMC6207163 DOI: 10.1089/ars.2017.7423] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Head and neck squamous cell cancer (HNSCC) is a complex disease characterized by high genetic and metabolic heterogeneity. Radiation therapy (RT) alone or combined with systemic chemotherapy is widely used for treatment of HNSCC as definitive treatment or as adjuvant treatment after surgery. Antibodies against epidermal growth factor receptor are used in definitive or palliative treatment. Recent Advances: Emerging targeted therapies against other proteins of interest as well as programmed cell death protein 1 and programmed death-ligand 1 immunotherapies are being explored in clinical trials. CRITICAL ISSUES The disease heterogeneity, invasiveness, and resistance to standard of care RT or chemoradiation therapy continue to constitute significant roadblocks for treatment and patients' quality of life (QOL) despite improvements in treatment modality and the emergence of new therapies over the past two decades. FUTURE DIRECTIONS As reviewed here, alterations in redox metabolism occur at all stages of HNSCC management, providing opportunities for improved prevention, early detection, response to therapies, and QOL. Bioinformatics and computational systems biology approaches are key to integrate redox effects with multiomics data from cells and clinical specimens and to identify redox modifiers or modifiable target proteins to achieve improved clinical outcomes. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jade Mims
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiumei Huang
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Naveen Singh
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Edward Motea
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | | | - Muhammad Beg
- Department of Internal Medicine, Division of Hematology-Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Melissa L. Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - David A. Boothman
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
127
|
El-Mohtadi F, d'Arcy R, Tirelli N. Oxidation-Responsive Materials: Biological Rationale, State of the Art, Multiple Responsiveness, and Open Issues. Macromol Rapid Commun 2018; 40:e1800699. [DOI: 10.1002/marc.201800699] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/13/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Farah El-Mohtadi
- Division of Pharmacy and Optometry; School of Health Sciences; Faculty of Biology; Medicine, and Health; The University of Manchester; Manchester M13 9PT UK
| | - Richard d'Arcy
- Laboratory of Polymers and Biomaterials; Fondazione Istituto Italiano di Tecnologia; 16163 Genova Italy
| | - Nicola Tirelli
- Division of Pharmacy and Optometry; School of Health Sciences; Faculty of Biology; Medicine, and Health; The University of Manchester; Manchester M13 9PT UK
- Laboratory of Polymers and Biomaterials; Fondazione Istituto Italiano di Tecnologia; 16163 Genova Italy
| |
Collapse
|
128
|
Nordzieke DE, Medraño-Fernandez I. The Plasma Membrane: A Platform for Intra- and Intercellular Redox Signaling. Antioxidants (Basel) 2018; 7:antiox7110168. [PMID: 30463362 PMCID: PMC6262572 DOI: 10.3390/antiox7110168] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023] Open
Abstract
Membranes are of outmost importance to allow for specific signal transduction due to their ability to localize, amplify, and direct signals. However, due to the double-edged nature of reactive oxygen species (ROS)—toxic at high concentrations but essential signal molecules—subcellular localization of ROS-producing systems to the plasma membrane has been traditionally regarded as a protective strategy to defend cells from unwanted side-effects. Nevertheless, specialized regions, such as lipid rafts and caveolae, house and regulate the activated/inhibited states of important ROS-producing systems and concentrate redox targets, demonstrating that plasma membrane functions may go beyond acting as a securing lipid barrier. This is nicely evinced by nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases (NOX), enzymes whose primary function is to generate ROS and which have been shown to reside in specific lipid compartments. In addition, membrane-inserted bidirectional H2O2-transporters modulate their conductance precisely during the passage of the molecules through the lipid bilayer, ensuring time-scaled delivery of the signal. This review aims to summarize current evidence supporting the role of the plasma membrane as an organizing center that serves as a platform for redox signal transmission, particularly NOX-driven, providing specificity at the same time that limits undesirable oxidative damage in case of malfunction. As an example of malfunction, we explore several pathological situations in which an inflammatory component is present, such as inflammatory bowel disease and neurodegenerative disorders, to illustrate how dysregulation of plasma-membrane-localized redox signaling impacts normal cell physiology.
Collapse
Affiliation(s)
- Daniela E Nordzieke
- Institute of Microbiology and Genetics, Department of Genetics of Eukaryotic Microorganisms, Georg August University Göttingen, Grisebachstr. 8, D-37077 Göttingen, Germany.
| | - Iria Medraño-Fernandez
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
129
|
Kostyuk AI, Panova AS, Bilan DS, Belousov VV. Redox biosensors in a context of multiparameter imaging. Free Radic Biol Med 2018; 128:23-39. [PMID: 29630928 DOI: 10.1016/j.freeradbiomed.2018.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/18/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022]
Abstract
A wide variety of genetically encoded fluorescent biosensors are available to date. Some of them have already contributed significantly to our understanding of biological processes occurring at cellular and organismal levels. Using such an approach, outstanding success has been achieved in the field of redox biology. The probes allowed researchers to observe, for the first time, the dynamics of important redox parameters in vivo during embryogenesis, aging, the inflammatory response, the pathogenesis of various diseases, and many other processes. Given the differences in the readout and spectra of the probes, they can be used in multiparameter imaging in which several processes are monitored simultaneously in the cell. Intracellular processes form an extensive network of interactions. For example, redox changes are often accompanied by changes in many other biochemical reactions related to cellular metabolism and signaling. Therefore, multiparameter imaging can provide important information concerning the temporal and spatial relationship of various signaling and metabolic processes. In this review, we will describe the main types of genetically encoded biosensors, the most frequently used readout, and their use in multiplexed imaging mode.
Collapse
Affiliation(s)
- Alexander I Kostyuk
- Faculty of Biology, Moscow State University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Anastasiya S Panova
- Faculty of Biology, Moscow State University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Dmitry S Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vsevolod V Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; Pirogov Russian National Research Medical University, Moscow 117997, Russia; Institute for Cardiovascular Physiology, Georg August University Göttingen, Göttingen D-37073, Germany.
| |
Collapse
|
130
|
Akter S, Fu L, Jung Y, Conte ML, Lawson JR, Lowther WT, Sun R, Liu K, Yang J, Carroll KS. Chemical proteomics reveals new targets of cysteine sulfinic acid reductase. Nat Chem Biol 2018; 14:995-1004. [PMID: 30177848 PMCID: PMC6192846 DOI: 10.1038/s41589-018-0116-2] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/18/2018] [Indexed: 01/06/2023]
Abstract
Cysteine sulfinic acid or S-sulfinylation is an oxidative post-translational modification (OxiPTM) that is known to be involved in redox-dependent regulation of protein function but has been historically difficult to analyze biochemically. To facilitate the detection of S-sulfinylated proteins, we demonstrate that a clickable, electrophilic diazene probe (DiaAlk) enables capture and site-centric proteomic analysis of this OxiPTM. Using this workflow, we revealed a striking difference between sulfenic acid modification (S-sulfenylation) and the S-sulfinylation dynamic response to oxidative stress, which is indicative of different roles for these OxiPTMs in redox regulation. We also identified >55 heretofore-unknown protein substrates of the cysteine sulfinic acid reductase sulfiredoxin, extending its function well beyond those of 2-cysteine peroxiredoxins (2-Cys PRDX1-4) and offering new insights into the role of this unique oxidoreductase as a central mediator of reactive oxygen species-associated diseases, particularly cancer. DiaAlk therefore provides a novel tool to profile S-sulfinylated proteins and study their regulatory mechanisms in cells.
Collapse
Affiliation(s)
- Salma Akter
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Youngeun Jung
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Mauro Lo Conte
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
- Novo Nordisk Research Center, Seattle, WA, USA
| | - J Reed Lawson
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - W Todd Lowther
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Rui Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Keke Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China.
- National Center for Protein Sciences-Beijing, Beijing, China.
- Beijing Institute of Lifeomics, Beijing, China.
| | - Kate S Carroll
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
131
|
Heppner DE, Dustin CM, Liao C, Hristova M, Veith C, Little AC, Ahlers BA, White SL, Deng B, Lam YW, Li J, van der Vliet A. Direct cysteine sulfenylation drives activation of the Src kinase. Nat Commun 2018; 9:4522. [PMID: 30375386 PMCID: PMC6207713 DOI: 10.1038/s41467-018-06790-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/19/2018] [Indexed: 01/17/2023] Open
Abstract
The Src kinase controls aspects of cell biology and its activity is regulated by intramolecular structural changes induced by protein interactions and tyrosine phosphorylation. Recent studies indicate that Src is additionally regulated by redox-dependent mechanisms, involving oxidative modification(s) of cysteines within the Src protein, although the nature and molecular-level impact of Src cysteine oxidation are unknown. Using a combination of biochemical and cell-based studies, we establish the critical importance of two Src cysteine residues, Cys-185 and Cys-277, as targets for H2O2-mediated sulfenylation (Cys-SOH) in redox-dependent kinase activation in response to NADPH oxidase-dependent signaling. Molecular dynamics and metadynamics simulations reveal the structural impact of sulfenylation of these cysteines, indicating that Cys-277-SOH enables solvent exposure of Tyr-416 to promote its (auto)phosphorylation, and that Cys-185-SOH destabilizes pTyr-527 binding to the SH2 domain. These redox-dependent Src activation mechanisms offer opportunities for development of Src-selective inhibitors in treatment of diseases where Src is aberrantly activated.
Collapse
Affiliation(s)
- David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA.
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Chenyi Liao
- Department of Chemistry, College of Arts and Sciences, University of Vermont, 82 University Place, Burlington, VT, 05405, USA
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Carmen Veith
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Bethany A Ahlers
- Department of Biology, College of Arts and Sciences, University of Vermont, 109 Carrigan Drive, Burlington, VT, 05405, USA
| | - Sheryl L White
- Department of Neurological Sciences, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Bin Deng
- Department of Biology, College of Arts and Sciences, University of Vermont, 109 Carrigan Drive, Burlington, VT, 05405, USA
| | - Ying-Wai Lam
- Department of Biology, College of Arts and Sciences, University of Vermont, 109 Carrigan Drive, Burlington, VT, 05405, USA
| | - Jianing Li
- Department of Chemistry, College of Arts and Sciences, University of Vermont, 82 University Place, Burlington, VT, 05405, USA.
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine University of Vermont, 149 Beaumont Avenue, Burlington, VT, 05405, USA.
| |
Collapse
|
132
|
Peng H, Zhang Y, Trinidad JC, Giedroc DP. Thioredoxin Profiling of Multiple Thioredoxin-Like Proteins in Staphylococcus aureus. Front Microbiol 2018; 9:2385. [PMID: 30374335 PMCID: PMC6196236 DOI: 10.3389/fmicb.2018.02385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/18/2018] [Indexed: 12/23/2022] Open
Abstract
Hydrogen sulfide (H2S) is thought to signal through protein S-sulfuration (persulfidation; S-sulfhydration) in both mammalian systems and bacteria. We previously profiled proteome S-sulfuration in Staphylococcus aureus (S. aureus) and identified two thioredoxin-like proteins, designated TrxP and TrxQ, that were capable of reducing protein persulfides as a potential regulatory mechanism. In this study, we further characterize TrxP, TrxQ and the canonical thioredoxin, TrxA, by identifying candidate protein substrates in S. aureus cells using a mechanism-based profiling assay where we trap mixed disulfides that exist between the attacking cysteine of a FLAG-tagged Trx and a persulfidated cysteine on the candidate substrate protein in cells. Largely non-overlapping sets of four, 32 and three candidate cellular substrates were detected for TrxA, TrxP, and TrxQ, respectively, many of which were previously identified as global proteome S-sulfuration targets including for example, pyruvate kinase, PykA. Both TrxA (k cat = 0.13 s-1) and TrxP (k cat = 0.088 s-1) are capable of reducing protein persulfides on PykA, a model substrate detected as a candidate substrate of TrxP; in contrast, TrxQ shows lower activity (k cat = 0.015 s-1). This work reveals that protein S-sulfuration, central to H2S and reactive sulfur species (RSS) signaling, may impact cellular activities and appears to be regulated in S. aureus largely by TrxP under conditions of sulfide stress.
Collapse
Affiliation(s)
- Hui Peng
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States.,Biochemistry Graduate Program, Indiana University Bloomington, Bloomington, IN, United States
| | - Yixiang Zhang
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States.,Laboratory for Biological Mass Spectrometry, Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States
| | - Jonathan C Trinidad
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States.,Laboratory for Biological Mass Spectrometry, Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States
| | - David P Giedroc
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States.,Department of Molecular and Cellular Biochemistry, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
133
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
134
|
Abstract
The abundance of oxidants and reductants must be balanced for an organism to thrive. Bacteria have evolved methods to prevent redox imbalances and to mitigate their deleterious consequences through the expression of detoxification enzymes, antioxidants, and systems to repair or degrade damaged proteins and DNA. Regulating these processes in response to redox changes requires sophisticated surveillance strategies ranging from metal chelation to direct sensing of toxic reactive oxygen species. In the case of bacterial pathogens, stress that threatens to disrupt redox homeostasis can derive from endogenous sources (produced by the bacteria) or exogenous sources (produced by the host). This minireview summarizes the sources of redox stress encountered during infection, the mechanisms by which bacterial pathogens diminish the damaging effects of redox stress, and the clever ways some organisms have evolved to thrive in the face of redox challenges during infection.
Collapse
|
135
|
Fashioning blood vessels by ROS signalling and metabolism. Semin Cell Dev Biol 2018; 80:35-42. [DOI: 10.1016/j.semcdb.2017.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/24/2017] [Accepted: 08/01/2017] [Indexed: 12/16/2022]
|
136
|
Foyer CH, Wilson MH, Wright MH. Redox regulation of cell proliferation: Bioinformatics and redox proteomics approaches to identify redox-sensitive cell cycle regulators. Free Radic Biol Med 2018; 122:137-149. [PMID: 29605447 PMCID: PMC6146653 DOI: 10.1016/j.freeradbiomed.2018.03.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/16/2018] [Accepted: 03/27/2018] [Indexed: 01/16/2023]
Abstract
Plant stem cells are the foundation of plant growth and development. The balance of quiescence and division is highly regulated, while ensuring that proliferating cells are protected from the adverse effects of environment fluctuations that may damage the genome. Redox regulation is important in both the activation of proliferation and arrest of the cell cycle upon perception of environmental stress. Within this context, reactive oxygen species serve as 'pro-life' signals with positive roles in the regulation of the cell cycle and survival. However, very little is known about the metabolic mechanisms and redox-sensitive proteins that influence cell cycle progression. We have identified cysteine residues on known cell cycle regulators in Arabidopsis that are potentially accessible, and could play a role in redox regulation, based on secondary structure and solvent accessibility likelihoods for each protein. We propose that redox regulation may function alongside other known posttranslational modifications to control the functions of core cell cycle regulators such as the retinoblastoma protein. Since our current understanding of how redox regulation is involved in cell cycle control is hindered by a lack of knowledge regarding both which residues are important and how modification of those residues alters protein function, we discuss how critical redox modifications can be mapped at the molecular level.
Collapse
Affiliation(s)
- Christine H Foyer
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| | - Michael H Wilson
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Megan H Wright
- The Astbury Centre for Structural Molecular Biology, School of Chemistry, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
137
|
Abstract
SIGNIFICANCE Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth. Recent Advances: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance. CRITICAL ISSUES The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes. FUTURE DIRECTIONS The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling. Antioxid. Redox Signal. 28, 1603-1625.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
138
|
Fassler R, Edinger N, Rimon O, Reichmann D. Defining Hsp33's Redox-regulated Chaperone Activity and Mapping Conformational Changes on Hsp33 Using Hydrogen-deuterium Exchange Mass Spectrometry. J Vis Exp 2018. [PMID: 29939186 DOI: 10.3791/57806] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Living organisms regularly need to cope with fluctuating environments during their life cycle, including changes in temperature, pH, the accumulation of reactive oxygen species, and more. These fluctuations can lead to a widespread protein unfolding, aggregation, and cell death. Therefore, cells have evolved a dynamic and stress-specific network of molecular chaperones, which maintain a "healthy" proteome during stress conditions. ATP-independent chaperones constitute one major class of molecular chaperones, which serve as first-line defense molecules, protecting against protein aggregation in a stress-dependent manner. One feature these chaperones have in common is their ability to utilize structural plasticity for their stress-specific activation, recognition, and release of the misfolded client. In this paper, we focus on the functional and structural analysis of one such intrinsically disordered chaperone, the bacterial redox-regulated Hsp33, which protects proteins against aggregation during oxidative stress. Here, we present a toolbox of diverse techniques for studying redox-regulated chaperone activity, as well as for mapping conformational changes of the chaperone, underlying its activity. Specifically, we describe a workflow which includes the preparation of fully reduced and fully oxidized proteins, followed by an analysis of the chaperone anti-aggregation activity in vitro using light-scattering, focusing on the degree of the anti-aggregation activity and its kinetics. To overcome frequent outliers accumulated during aggregation assays, we describe the usage of Kfits, a novel graphical tool which allows easy processing of kinetic measurements. This tool can be easily applied to other types of kinetic measurements for removing outliers and fitting kinetic parameters. To correlate the function with the protein structure, we describe the setup and workflow of a structural mass spectrometry technique, hydrogen-deuterium exchange mass spectrometry, that allows the mapping of conformational changes on the chaperone and substrate during different stages of Hsp33 activity. The same methodology can be applied to other protein-protein and protein-ligand interactions.
Collapse
Affiliation(s)
- Rosi Fassler
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem
| | - Nufar Edinger
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem
| | - Oded Rimon
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem
| | - Dana Reichmann
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem;
| |
Collapse
|
139
|
Fukuto JM, Ignarro LJ, Nagy P, Wink DA, Kevil CG, Feelisch M, Cortese-Krott MM, Bianco CL, Kumagai Y, Hobbs AJ, Lin J, Ida T, Akaike T. Biological hydropersulfides and related polysulfides - a new concept and perspective in redox biology. FEBS Lett 2018; 592:2140-2152. [PMID: 29754415 PMCID: PMC6033183 DOI: 10.1002/1873-3468.13090] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
Abstract
The chemical biology of thiols (RSH, e.g., cysteine and cysteine-containing proteins/peptides) has been a topic of extreme interest for many decades due to their reported roles in protein structure/folding, redox signaling, metal ligation, cellular protection, and enzymology. While many of the studies on thiol/sulfur biochemistry have focused on thiols, relatively ignored have been hydropersulfides (RSSH) and higher order polysulfur species (RSSn H, RSSn R, n > 1). Recent and provocative work has alluded to the prevalence and likely physiological importance of RSSH and related RSSn H. RSSH of cysteine (Cys-SSH) has been found to be prevalent in mammalian systems along with Cys-SSH-containing proteins. The RSSH functionality has not been examined to the extent of other biologically relevant sulfur derivatives (e.g., sulfenic acids, disulfides, etc.), whose roles in cell signaling are strongly indicated. The recent finding of Cys-SSH biosynthesis and translational incorporation into proteins is an unequivocal indication of its fundamental importance and necessitates a more profound look into the physiology of RSSH. In this Review, we discuss the currently reported chemical biology of RSSH (and related species) as a prelude to discussing their possible physiological roles.
Collapse
Affiliation(s)
- Jon M Fukuto
- Department of Chemistry, Sonoma State University, Rohnert Park, CA, USA
| | - Louis J Ignarro
- Department of Molecular and Medical Pharmacology, Center for the Health Sciences, UCLA School of Medicine, Los Angeles, CA, USA
| | - Peter Nagy
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | - David A Wink
- Tumor Biology Section, Radiation Biology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana Statue University Health Sciences Center, Shreveport, LA, USA
| | - Martin Feelisch
- NIHR Southampton Biomedical Research Center, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany
| | - Christopher L Bianco
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany
| | - Yoshito Kumagai
- Environmental Biology Section, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Adrian J Hobbs
- William Harvey Research Institute, Bart & London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| | - Joseph Lin
- Department of Biology, Sonoma State University, Rohnert Park, CA, USA
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
140
|
Glanville DG, Han L, Maule AF, Woodacre A, Thanki D, Abdullah IT, Morrissey JA, Clarke TB, Yesilkaya H, Silvaggi NR, Ulijasz AT. RitR is an archetype for a novel family of redox sensors in the streptococci that has evolved from two-component response regulators and is required for pneumococcal colonization. PLoS Pathog 2018; 14:e1007052. [PMID: 29750817 PMCID: PMC5965902 DOI: 10.1371/journal.ppat.1007052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/23/2018] [Accepted: 04/23/2018] [Indexed: 01/07/2023] Open
Abstract
To survive diverse host environments, the human pathogen Streptococcus pneumoniae must prevent its self-produced, extremely high levels of peroxide from reacting with intracellular iron. However, the regulatory mechanism(s) by which the pneumococcus accomplishes this balance remains largely enigmatic, as this pathogen and other related streptococci lack all known redox-sensing transcription factors. Here we describe a two-component-derived response regulator, RitR, as the archetype for a novel family of redox sensors in a subset of streptococcal species. We show that RitR works to both repress iron transport and enable nasopharyngeal colonization through a mechanism that exploits a single cysteine (Cys128) redox switch located within its linker domain. Biochemical experiments and phylogenetics reveal that RitR has diverged from the canonical two-component virulence regulator CovR to instead dimerize and bind DNA only upon Cys128 oxidation in air-rich environments. Atomic structures show that Cys128 oxidation initiates a "helical unravelling" of the RitR linker region, suggesting a mechanism by which the DNA-binding domain is then released to interact with its cognate regulatory DNA. Expanded computational studies indicate this mechanism could be shared by many microbial species outside the streptococcus genus.
Collapse
Affiliation(s)
- David G. Glanville
- Department of Microbiology and Immunology, Loyola University Chicago; Maywood, IL, United States of America
- MRC Centre for Molecular Bacteriology and Infection (CMBI), Imperial College London, London, United Kingdom
| | - Lanlan Han
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Andrew F. Maule
- Department of Horticulture, University of Wisconsin–Madison, Linden Drive, Madison, Wisconsin, United States of America
| | - Alexandra Woodacre
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Devsaagar Thanki
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Iman Tajer Abdullah
- Department of Infection and Immunity, University of Leicester, Leicester, United Kingdom
| | - Julie A. Morrissey
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Thomas B. Clarke
- MRC Centre for Molecular Bacteriology and Infection (CMBI), Imperial College London, London, United Kingdom
| | - Hasan Yesilkaya
- Department of Infection and Immunity, University of Leicester, Leicester, United Kingdom
| | - Nicholas R. Silvaggi
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Andrew T. Ulijasz
- Department of Microbiology and Immunology, Loyola University Chicago; Maywood, IL, United States of America
- MRC Centre for Molecular Bacteriology and Infection (CMBI), Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
141
|
Goemans CV, Vertommen D, Agrebi R, Collet JF. CnoX Is a Chaperedoxin: A Holdase that Protects Its Substrates from Irreversible Oxidation. Mol Cell 2018; 70:614-627.e7. [PMID: 29754824 DOI: 10.1016/j.molcel.2018.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/22/2018] [Accepted: 04/03/2018] [Indexed: 02/01/2023]
Abstract
Bleach (HOCl) is a powerful oxidant that kills bacteria in part by causing protein aggregation. It inactivates ATP-dependent chaperones, rendering cellular proteins mostly dependent on holdases. Here we identified Escherichia coli CnoX (YbbN) as a folding factor that, when activated by bleach via chlorination, functions as an efficient holdase, protecting the substrates of the major folding systems GroEL/ES and DnaK/J/GrpE. Remarkably, CnoX uniquely combines this function with the ability to prevent the irreversible oxidation of its substrates. This dual activity makes CnoX the founding member of a family of proteins, the "chaperedoxins." Because CnoX displays a thioredoxin fold and a tetratricopeptide (TPR) domain, two structural motifs conserved in all organisms, this investigation sets the stage for the discovery of additional chaperedoxins in bacteria and eukaryotes that could cooperate with proteins from both the Hsp60 and Hsp70 families.
Collapse
Affiliation(s)
- Camille V Goemans
- WELBIO, Avenue Hippocrate 75, 1200 Brussels, Belgium; de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, 1200 Brussels, Belgium
| | - Didier Vertommen
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, 1200 Brussels, Belgium
| | - Rym Agrebi
- WELBIO, Avenue Hippocrate 75, 1200 Brussels, Belgium; de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, 1200 Brussels, Belgium
| | - Jean-François Collet
- WELBIO, Avenue Hippocrate 75, 1200 Brussels, Belgium; de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| |
Collapse
|
142
|
Bestetti S, Medraño-Fernandez I, Galli M, Ghitti M, Bienert GP, Musco G, Orsi A, Rubartelli A, Sitia R. A persulfidation-based mechanism controls aquaporin-8 conductance. SCIENCE ADVANCES 2018; 4:eaar5770. [PMID: 29732408 PMCID: PMC5931763 DOI: 10.1126/sciadv.aar5770] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/16/2018] [Indexed: 05/20/2023]
Abstract
Upon engagement of tyrosine kinase receptors, nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases release H2O2 in the extracellular space. We reported previously that aquaporin-8 (AQP8) transports H2O2 across the plasma membrane and is reversibly gated during cell stress, modulating signal strength and duration. We show that AQP8 gating is mediated by persulfidation of cysteine 53 (C53). Treatment with H2S is sufficient to block H2O2 entry in unstressed cells. Silencing cystathionine β-synthase (CBS) prevents closure, suggesting that this enzyme is the main source of H2S. Molecular modeling indicates that C53 persulfidation displaces a nearby histidine located in the narrowest part of the channel. We propose that H2O2 molecules transported through AQP8 sulfenylate C53, making it susceptible to H2S produced by CBS. This mechanism tunes H2O2 transport and may control signaling and limit oxidative stress.
Collapse
Affiliation(s)
- Stefano Bestetti
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Iria Medraño-Fernandez
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Corresponding author. (R.S.); (I.M.-F.)
| | - Mauro Galli
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Michela Ghitti
- Biomolecular Nuclear Magnetic Resonance (NMR) Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Gerd P. Bienert
- Metalloid Transport Group, Leibniz Institute of Plant Genetics and Crop Plant Research, 06466 Gatersleben, Germany
| | - Giovanna Musco
- Biomolecular Nuclear Magnetic Resonance (NMR) Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Andrea Orsi
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Azienda Ospedaliera Universitaria (AOU) San Martino-IST, 16132 Genoa, Italy
| | - Roberto Sitia
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Corresponding author. (R.S.); (I.M.-F.)
| |
Collapse
|
143
|
Genetic Code Expansion: A Powerful Tool for Understanding the Physiological Consequences of Oxidative Stress Protein Modifications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7607463. [PMID: 29849913 PMCID: PMC5937447 DOI: 10.1155/2018/7607463] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/19/2018] [Indexed: 12/17/2022]
Abstract
Posttranslational modifications resulting from oxidation of proteins (Ox-PTMs) are present intracellularly under conditions of oxidative stress as well as basal conditions. In the past, these modifications were thought to be generic protein damage, but it has become increasingly clear that Ox-PTMs can have specific physiological effects. It is an arduous task to distinguish between the two cases, as multiple Ox-PTMs occur simultaneously on the same protein, convoluting analysis. Genetic code expansion (GCE) has emerged as a powerful tool to overcome this challenge as it allows for the site-specific incorporation of an Ox-PTM into translated protein. The resulting homogeneously modified protein products can then be rigorously characterized for the effects of individual Ox-PTMs. We outline the strengths and weaknesses of GCE as they relate to the field of oxidative stress and Ox-PTMs. An overview of the Ox-PTMs that have been genetically encoded and applications of GCE to the study of Ox-PTMs, including antibody validation and therapeutic development, is described.
Collapse
|
144
|
Samet JM, Wages PA. Oxidative Stress from Environmental Exposures. CURRENT OPINION IN TOXICOLOGY 2018; 7:60-66. [PMID: 30079382 PMCID: PMC6069528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Oxidative stress is arguably the most common mechanism in the toxicology of environmental agents, unifying the action of broad classes of physichochemically disparate environmental pollutants, including oxidant gases, organic compounds, particulate surfaces, and metal ions. As advances in redox biology identify previously unrecognized targets for disruption by exposure to xenobiotics, redox toxicology has emerged as a new field of investigation. Environmental contaminants can induce oxidative stress on cells through mechanisms that are direct, indirect or involve the disruption of metabolic or bioenergetic processes that are regulated by thiol redox switches. Live-cell imaging has proven to be a powerful approach to the study of environmental oxidative stress. Cells are equipped with multiple complementary energy-dependent systems for maintaining redox homeostasis in the face of environmental oxidative stress.
Collapse
Affiliation(s)
- James M. Samet
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27707
| | - Phillip A. Wages
- Department of Chemistry, Vanderbilt University, Nashville, TN 49795
| |
Collapse
|
145
|
Virreira Winter S, Zychlinsky A. The bacterial pigment pyocyanin inhibits the NLRP3 inflammasome through intracellular reactive oxygen and nitrogen species. J Biol Chem 2018; 293:4893-4900. [PMID: 29414783 PMCID: PMC5880120 DOI: 10.1074/jbc.ra117.001105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/05/2018] [Indexed: 01/10/2023] Open
Abstract
Inflammasomes are cytosolic complexes that mature and secrete the inflammatory cytokines interleukin 1β (IL-1β) and IL-18 and induce pyroptosis. The NLRP3 (NACHT, LRR, and PYD domains–containing protein 3) inflammasome detects many pathogen- and danger-associated molecular patterns, and reactive oxygen species (ROS)/reactive nitrogen species (RNS) have been implicated in its activation. The phenazine pyocyanin (PCN) is a virulence factor of Pseudomonas aeruginosa and generates superoxide in cells. Here we report that PCN inhibits IL-1β and IL-18 release and pyroptosis upon NLRP3 inflammasome activation in macrophages by preventing speck formation and Caspase-1 maturation. Of note, PCN did not regulate the AIM2 (absent in melanoma 2) or NLRC4 inflammasomes or tumor necrosis factor (TNF) secretion. Imaging of the fluorescent glutathione redox potential sensor Grx1-roGFP2 indicated that PCN provokes cytosolic and nuclear but not mitochondrial redox changes. PCN-induced intracellular ROS/RNS inhibited the NLRP3 inflammasome posttranslationally, and hydrogen peroxide or peroxynitrite alone were sufficient to block its activation. We propose that cytosolic ROS/RNS inhibit the NLRP3 inflammasome and that PCN's anti-inflammatory activity may help P. aeruginosa evade immune recognition.
Collapse
Affiliation(s)
| | - Arturo Zychlinsky
- Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
146
|
|
147
|
Gopinath P, Ohayon S, Nawatha M, Brik A. Chemical and semisynthetic approaches to study and target deubiquitinases. Chem Soc Rev 2018; 45:4171-98. [PMID: 27049734 DOI: 10.1039/c6cs00083e] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ubiquitination is a key posttranslational modification, which affects numerous biological processes and is reversed by a class of enzymes known as deubiquitinases (DUBs). This family of enzymes cleaves mono-ubiquitin or poly-ubiquitin chains from a target protein through different mechanisms and mode of interactions with their substrates. Studying the role of DUBs in health and diseases has been a major goal for many laboratories both in academia and in industry. However, the field has been challenged by the difficulties in obtaining native substrates and novel reagents using traditional enzymatic and molecular biology approaches. Recent advancements in the synthesis and semisynthesis of proteins made it possible to prepare several unique ubiquitin conjugates to study various aspects of DUBs such as their specificities and structures. Moreover, these approaches enable the preparation of novel activity based probes and assays to monitor DUB activities in vitro and in cellular contexts. Efforts made to bring new chemical entities for the selective inhibition of DUBs based on these tools are also highlighted with selected examples.
Collapse
Affiliation(s)
- Pushparathinam Gopinath
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology Haifa, 3200008, Israel.
| | - Shimrit Ohayon
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology Haifa, 3200008, Israel.
| | - Mickal Nawatha
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology Haifa, 3200008, Israel.
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology Haifa, 3200008, Israel.
| |
Collapse
|
148
|
Ethanol Induced Disordering of Pancreatic Acinar Cell Endoplasmic Reticulum: An ER Stress/Defective Unfolded Protein Response Model. Cell Mol Gastroenterol Hepatol 2018; 5:479-497. [PMID: 29930975 PMCID: PMC6009017 DOI: 10.1016/j.jcmgh.2018.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/02/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Heavy alcohol drinking is associated with pancreatitis, whereas moderate intake lowers the risk. Mice fed ethanol long term show no pancreas damage unless adaptive/protective responses mediating proteostasis are disrupted. Pancreatic acini synthesize digestive enzymes (largely serine hydrolases) in the endoplasmic reticulum (ER), where perturbations (eg, alcohol consumption) activate adaptive unfolded protein responses orchestrated by spliced X-box binding protein 1 (XBP1). Here, we examined ethanol-induced early structural changes in pancreatic ER proteins. METHODS Wild-type and Xbp1+/- mice were fed control and ethanol diets, then tissues were homogenized and fractionated. ER proteins were labeled with a cysteine-reactive probe, isotope-coded affinity tag to obtain a novel pancreatic redox ER proteome. Specific labeling of active serine hydrolases in ER with fluorophosphonate desthiobiotin also was characterized proteomically. Protein structural perturbation by redox changes was evaluated further in molecular dynamic simulations. RESULTS Ethanol feeding and Xbp1 genetic inhibition altered ER redox balance and destabilized key proteins. Proteomic data and molecular dynamic simulations of Carboxyl ester lipase (Cel), a unique serine hydrolase active within ER, showed an uncoupled disulfide bond involving Cel Cys266, Cel dimerization, ER retention, and complex formation in ethanol-fed, XBP1-deficient mice. CONCLUSIONS Results documented in ethanol-fed mice lacking sufficient spliced XBP1 illustrate consequences of ER stress extended by preventing unfolded protein response from fully restoring pancreatic acinar cell proteostasis during ethanol-induced redox challenge. In this model, orderly protein folding and transport to the secretory pathway were disrupted, and abundant molecules including Cel with perturbed structures were retained in ER, promoting ER stress-related pancreas pathology.
Collapse
Key Words
- %-ox, percentage oxidized
- ATPase, adenosine triphosphatase
- Alcohol Pancreatitis
- Carboxyl Ester Lipase
- Cel, carboxyl ester lipase
- DTT, dithiothreitol
- Disulfide Bond
- ER, endoplasmic reticulum
- ERAD, endoplasmic reticulum–associated degradation
- FAEE, fatty acid ethyl esters
- FP, fluorophosphonate
- ICAT, isotope-coded affinity tags
- LC-MS/MS, liquid chromatography-tandem mass spectrometry
- MW, molecular weight
- RER, rough ER
- UPR, unfolded protein response
- Unfolded Protein Response
- WT, wild type
- sXBP1, spliced X box-binding protein 1
Collapse
|
149
|
Alcock LJ, Perkins MV, Chalker JM. Chemical methods for mapping cysteine oxidation. Chem Soc Rev 2018; 47:231-268. [DOI: 10.1039/c7cs00607a] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Methods to characterise oxidative modifications of cysteine help clarify their role in protein function in both healthy and diseased cells.
Collapse
Affiliation(s)
- Lisa J. Alcock
- College of Science and Engineering
- Flinders University
- South Australia
- Australia
| | - Michael V. Perkins
- College of Science and Engineering
- Flinders University
- South Australia
- Australia
| | - Justin M. Chalker
- College of Science and Engineering
- Flinders University
- South Australia
- Australia
| |
Collapse
|
150
|
Abstract
Cysteine thiols are involved in a diverse set of biological transformations, including nucleophilic and redox catalysis, metal coordination and formation of both dynamic and structural disulfides. Often posttranslationally modified, cysteines are also frequently alkylated by electrophilic compounds, including electrophilic metabolites, drugs, and natural products, and are attractive sites for covalent probe and drug development. Quantitative proteomics combined with activity-based protein profiling has been applied to annotate cysteine reactivity, susceptibility to posttranslational modifications, and accessibility to chemical probes, uncovering thousands of functional and small-molecule targetable cysteines across a diverse set of proteins, proteome-wide in an unbiased manner. Reactive cysteines have been targeted by high-throughput screening and fragment-based ligand discovery efforts. New cysteine-reactive electrophiles and compound libraries have been synthesized to enable inhibitor discovery broadly and to minimize nonspecific toxicity and off-target activity of compounds. With the recent blockbuster success of several covalent inhibitors, and the development of new chemical proteomic strategies to broadly identify reactive, ligandable and posttranslationally modified cysteines, cysteine profiling is poised to enable the development of new potent and selective chemical probes and even, in some cases, new drugs.
Collapse
|